Next Article in Journal
Recent Advances in Pepper Fruit Glossiness
Previous Article in Journal
The Role of Fc-like Receptor 3 in the Pathophysiology of Rheumatoid Arthritis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Sex-Specific Molecular and Genomic Responses to Endocrine Disruptors in Aquatic Species: The Central Role of Vitellogenin

by
Faustina Barbara Cannea
1,
Cristina Porcu
2,
Maria Cristina Follesa
2 and
Alessandra Padiglia
1,*
1
Department of Life and Environmental Sciences (DiSVA), Biomedical Section, University of Cagliari, Cittadella Universitaria di Monserrato, Monserrato, 09042 Cagliari, Italy
2
Department of Life and Environmental Sciences (DiSVA), Marine Bioecology Section, University of Cagliari, Via T. Fiorelli 1, 09126 Cagliari, Italy
*
Author to whom correspondence should be addressed.
Genes 2025, 16(11), 1317; https://doi.org/10.3390/genes16111317
Submission received: 5 September 2025 / Revised: 2 October 2025 / Accepted: 16 October 2025 / Published: 2 November 2025
(This article belongs to the Section Animal Genetics and Genomics)

Abstract

Endocrine-disrupting chemicals (EDCs) are widespread contaminants that interfere with hormonal signaling and compromise reproductive success in aquatic organisms. Vitellogenin (VTG) is one of the most widely established biomarkers of estrogenic exposure, especially in males and juveniles. However, evidence from multi-omics studies indicates that VTG induction occurs within broader transcriptional and regulatory networks, involving genes such as cyp19a1 (aromatase), cyp1a (cytochrome P4501A), and other stress-responsive genes, underscoring the complexity of endocrine disruption. This review focuses on nuclear receptor isoforms, including estrogen receptor alpha (ERα), estrogen receptor beta (ERβ), and androgen receptor (AR) variants. We examine the diversification of vtg gene repertoires across teleost genomes and epigenetic mechanisms, such as DNA methylation and microRNAs, that modulate sex-dependent sensitivity. In addition, we discuss integrative approaches that combine VTG with transcriptomic, epigenetic, and histological endpoints. Within the Adverse Outcome Pathway (AOP) and weight-of-evidence (WoE) frameworks, these strategies provide mechanistic links between receptor activation and reproductive impairment. Finally, we outline future directions, focusing on the development of sex-specific biomarker panels, the integration of omics-based data with machine learning, and advances in ecogenomics. Embedding molecular responses into ecological and regulatory contexts will help bridge mechanistic insights with environmental relevance and support sustainability goals such as SDG 14 (Life Below Water).

1. Introduction

Aquatic ecosystems are increasingly threatened by EDCs, a diverse group of contaminants that interfere with hormonal signaling and homeostasis in wildlife [1,2]. Their presence in surface waters and sediments has been widely documented, together with their capacity to compromise reproduction, growth, and development in aquatic organisms [3,4,5].
An essential dimension of endocrine disruption research is the consideration of sex as a biological variable. Males and females differ in circulating hormone levels, reproductive physiology, receptor distribution, metabolic capacity, and epigenetic regulation [6,7,8]. These differences shape sex-dependent molecular responses, whereby EDCs trigger divergent transcriptional, epigenetic, and physiological effects [9,10]. This variability has profound implications for reproductive fitness and population resilience, underscoring the need to incorporate sex-based perspectives into ecotoxicological assessment [11,12].
Among molecular biomarkers, the yolk precursor protein VTG is the most established indicator of estrogenic activity [13,14]. Synthesized in the liver of oviparous females under natural conditions, VTG is normally absent in males and juveniles but becomes strongly induced by exposure to estrogenic EDCs [11,12]. This makes VTG a robust and sensitive biomarker, extensively applied in ecotoxicology through both protein-based assays (ELISA, Western blotting) and transcript-level analyses (qPCR, RNA-Seq) [15,16,17,18]. Nevertheless, interpretation requires careful consideration of sex, reproductive stage, species variability, and environmental context.
Although fish dominate the literature, evidence from other taxa, such as mollusks, crustaceans, and amphibians, confirms that endocrine disruption is a cross-phyla phenomenon [19,20,21,22,23]. These findings emphasize the need for biomarker strategies that extend beyond fish and incorporate alternative molecular endpoints. Recent advances in omics technologies reveal that EDC responses extend beyond VTG and involve nuclear receptors, steroidogenic enzymes, detoxification pathways, and stress-related genes [24,25,26]. Placing VTG within this broader multi-omics framework underscores its central role and highlights the need for integrative approaches to capture the complexity of endocrine disruption.
The aim of this review is to provide an integrated perspective on sex-specific molecular responses to EDCs in aquatic organisms, with particular emphasis on the ecotoxicological relevance of VTG and its integration into multi-omics approaches. Ultimately, this work seeks to advance ecotoxicological assessment and contribute to global sustainability efforts, particularly the United Nations Sustainable Development Goal (SDG) 14: Life Below Water [27].

Methodology

This review was conducted as a narrative synthesis of the scientific literature. Relevant studies were retrieved from PubMed, Scopus, and Web of Science using combinations of the following keywords: “endocrine-disrupting chemicals” OR “EDCs”, “sex-specific responses”, “vitellogenin” OR “VTG”, “multi-omics”, “fish”, “mollusks”, “crustaceans”, and “aquatic organisms”. The main search covered the period from 2000 to 2025 to capture both seminal and most recent contributions. However, earlier key studies of fundamental relevance (e.g., discovery of VTG as a biomarker, initial reports of EDC effects) were also included to provide historical and conceptual context. In total, approximately 1200 records were initially retrieved, 300 screened in detail, and 189 included in this review.

2. Endocrine Disruptors in Aquatic Environments

EDCs are recognized as priority pollutants due to their ability to interfere with hormonal signaling and reproduction in aquatic organisms [28,29]. Although fish are particularly sensitive because of their constant waterborne exposure, invertebrates such as bivalves and crustaceans also display endocrine-sensitive processes, including vitellogenin (VTG)-like protein synthesis and disrupted steroidogenic pathways [30]. EDCs originate from agriculture, industry, urbanization, and wastewater discharge, with most ecotoxicological research focusing on four main categories: pesticides, plasticizers, pharmaceuticals, and industrial contaminants.

2.1. Main Sources of EDCs

Representative examples illustrate the breadth of EDC impacts. Legacy pesticides such as dichlorodiphenyltrichloroethane (DDT) persist in sediments and retain estrogenic activity decades after their ban [31]. Atrazine, one of the most widely used herbicides, alters aromatase activity and disrupts sex steroid balance, leading to feminization in fish and amphibians [32,33].
Plasticizers such as bisphenol A (BPA) and nonylphenol bind estrogen receptors and induce VTG in male fish, with field studies linking nonylphenol exposure to intersex conditions in wild roach (Rutilus rutilus) populations downstream of wastewater treatment plants [34,35]. In mollusks, BPA disrupts embryonic development in Mytilus galloprovincialis and alters shell formation and serotonergic signaling [36], whereas induction of VTG-like proteins remains inconsistent, with some studies reporting biomarker potential and others finding no induction even under potent EE2 exposure [37].
Among synthetic estrogens, 17α-ethinylestradiol (EE2) is particularly concerning, being resistant to degradation and active at ng/L concentrations. A landmark whole-lake experiment in Canada demonstrated that chronic EE2 exposure caused the collapse of Pimephales promelas populations, with recovery only after exposure ceased [38,39]. More broadly, reviews emphasize the profound impacts of estrogenic EDCs on reproduction in wildlife and humans [40].
Industrial contaminants such as polychlorinated biphenyls (PCBs) and dioxins act mainly through activation of the aryl hydrocarbon receptor (AhR), disrupting reproduction and development [41], while heavy metals such as cadmium and mercury impair steroid biosynthesis in both fish and mollusks.
Per- and polyfluoroalkyl substances (PFAS) are another class of persistent contaminants that interfere with thyroid and reproductive hormone regulation in fish [42,43] and in mollusks, where experimental exposures demonstrated immune and endocrine-related effects [44,45].

2.2. Exposure Pathways in Aquatic Organisms

EDCs enter aquatic organisms through multiple routes, including gill diffusion, dermal absorption, and oral ingestion. Transporters such as solute carriers (SLCs) and ATP-binding cassette (ABC) efflux pumps regulate bioavailability, while the intestinal microbiota can regenerate active estrogens from conjugated forms [46,47,48,49]. Lipophilic compounds (e.g., PCBs, organochlorine pesticides) accumulate in liver and gonads, enabling maternal transfer via VTG [50,51]. In contrast, polar contaminants such as PFAS circulate bound to plasma proteins and persist through enterohepatic recirculation [48,52,53]. In invertebrates, the hepatopancreas serves as the main detoxification organ, expressing Phase I/II enzymes and ABC transporters [54,55,56,57,58]. Crustaceans accumulate contaminants via gills and diet, with subsequent disruption of the ecdysteroid–methyl farnesoate axis [59,60,61]. Bioaccumulation and trophic transfer enhance contaminant transfer, with bivalves and crustaceans acting both as sentinels and vectors within marine food webs [62,63].

2.3. Physiological and Reproductive Effects of EDCs

At the molecular level, EDCs act through multiple mechanisms, including receptor binding, interference with steroidogenic enzymes, and epigenetic modifications [64,65,66,67,68,69,70,71,72,73,74]. These perturbations cascade into intersex gonads, impaired spermatogenesis with reduced sperm motility, abnormal oogenesis, altered sex steroid ratios, secondary sexual trait modifications, and reduced fecundity, with well-documented cases in fish populations exposed to estrogenic effluents and EE2. Long-term surveys in UK rivers have linked the feminization of wild R. rutilus populations directly to wastewater effluents, providing evidence that spans from ER activation to vtg gene induction and histopathological alterations. Similarly, the whole-lake EE2 experiment in Canada showed that molecular and histological perturbations scaled up to a demographic collapse, with recovery occurring only after exposure ceased [38,75].
In addition to freshwater fish, evidence from marine species highlights comparable effects. In Atlantic cod (Gadus morhua), multi-omics analyses revealed hepatic reprogramming of lipid metabolism and immunity following EE2 exposure [76]. European flounder (Platichthys flesus) collected from polluted estuaries exhibited co-induction of VTG, choriogenins, and heat shock proteins, confirming field-level estrogenic impacts [77]. These findings underscore that endocrine disruption in marine fish parallels observations in freshwater taxa, with similar molecular-to-population cascades.
In bivalves, exposure to estrogenic and anti-androgenic compounds alters gametogenesis, steroid metabolism, and larval development. Transcriptomic studies in Mytilus edulis and M. galloprovincialis revealed perturbations in lipid metabolism, serotonin/prostaglandin signaling, and vitellogenesis [19,78]. However, induction of VTG-like proteins remains inconsistent across species, underscoring the limitations of VTG as a universal biomarker in invertebrates [19].
In crustaceans, pesticides and organotins disrupt reproduction by interfering with the ecdysteroid–methyl farnesoate (MF) axis through the EcR/RXR receptor complex. These effects manifest as impaired molting, abnormal vitellogenesis, and reduced fertility, as supported by evidence on vitellogenin-like proteins in invertebrates [79], reviews of endocrine disruption in crustaceans [80], updated studies on organotin toxicity [81], and mechanistic insights into endocrine regulation of reproduction [82]. EcR/RXR-mediated signaling is a known target of endocrine toxicants [83], and experimental exposure to methyl farnesoate (MF) and 20-hydroxyecdysone (20E) in Marsupenaeus japonicus larvae has further confirmed the vulnerability of these pathways to disruption [84]. Moreover, crustacean endocrine physiology differs substantially from vertebrates, requiring taxon-specific evaluation of biomarkers [85].
Overall, these findings highlight the need for integrative biomarker approaches combining VTG, steroidogenic enzymes, receptor transcripts, and epigenetic endpoints. Within the Adverse Outcome Pathway (AOP) framework, such biomarkers can be linked to higher-level outcomes, thereby supporting regulatory risk assessment [86,87]. Collectively, they provide a comparative perspective across phyla, showing how endocrine-disrupting chemicals impact aquatic organisms at multiple levels of biological organization (Table 1).

3. Sex-Specific Molecular Responses to EDCs

The molecular effects of EDCs are strongly sex-dependent, reflecting dimorphisms in endocrine physiology, receptor isoform composition, gene repertoires, and epigenetic control. These differences lead to divergent transcriptional, epigenetic, and metabolic responses in males and females exposed to the same contaminant. Understanding such pathways is essential for interpreting biomarkers, refining ecotoxicological assessments, and developing predictive strategies aligned with genomics-based strategies.

3.1. Nuclear Receptor Isoforms (ERα, ERβ, and AR Variants)

Estrogen and androgen receptors are primary mediators of endocrine disruption, and their isoform diversity underpins sex-specific responses. In teleosts, gene duplication produced multiple ER isoforms, including ERα (esr1) and two ERβ paralogs (esr2a, esr2b), which differ in tissue distribution and transcriptional roles [88,89]. Functional studies confirm that these isoforms are not redundant, as each fulfills distinct roles in reproduction and development.
In zebrafish (Danio rerio), esr1 knockout abolishes VTG synthesis and impairs ovarian maturation, whereas disruption of esr2a/b affects fertility, sexual differentiation, and gonadal morphology [90]. ERα is therefore the main driver of hepatic vtg induction, while ERβ isoforms modulate reproduction and development. ER isoforms also differ in ligand-binding affinities: ERα has high affinity for natural estrogens such as 17β-estradiol (E2), whereas ERβ paralogs often respond more strongly to synthetic estrogens such as EE2 or BPA [91]. In addition to nuclear ERs, membrane-associated receptors also play a role, with GPER (gper1) mediating rapid signaling and inhibiting oocyte maturation in D. rerio [92]. Androgen receptors have also diversified, with ARα and ARβ encoded by distinct genes. ARα is predominant in gonadal development, whereas ARβ contributes mainly to neural and behavioral traits [92].
Anti-androgenic contaminants strongly affect AR-regulated pathways: in stickleback (Gasterosteus aculeatus), suppression of the AR-dependent protein spiggin by flutamide or fungicides directly impairs male fertility [93]. Other receptors also participate in sex-specific responses. PGRMC1 and mPRα are required for normal oocyte maturation in D. rerio [94,95], while the glucocorticoid receptor responds to salinity and immune stressors in several teleosts, including large yellow croaker (Larimichthys crocea) [96]. Cross-talk further modulates effects. Activation of AhR by dioxin-like PCBs antagonizes ER-mediated transcription and reduces vtg induction in males [97,98].
Emerging contaminants such as PFAS also interact with these receptors, raising concern for their disruptive potential across taxa [99]. In invertebrates, BPA and nonylphenol induce vtg-like and ER-related transcripts in M. galloprovincialis, while in crustaceans organotins disrupt nuclear receptor-mediated control of vitellogenesis and molting [61,71].

3.2. Diversification of vtg Gene Repertoires in Teleosts

The vtg gene family exemplifies how genomic architecture shapes biomarker responses. Successive genome duplications in teleosts generated multiple paralogs with distinct regulatory profiles. In D. rerio, eight vtg genes are organized into three types: five type I (vtg1, 47), two type II (vtg2, 8), and one type III (vtg3) [100,101]. It should be noted that two different classification systems are used in the literature: the designation of type I–III VTGs refers to structural features and domain organization, whereas the clade nomenclature (Aa, Ab, C) reflects phylogenetic relationships among teleost paralogs. Functional validation using CRISPR–Cas9 demonstrated that both type I and type III paralogs play essential roles in reproduction and early development, with vtg1 and vtg3 knockouts severely impairing fertility, embryo survival, and gonadal differentiation [102]. In salmonids, which underwent an additional genome duplication, large vtg multigene arrays have been described, often organized in chromosomal clusters [103]. Some paralogs are highly estrogen-responsive, while others show minimal induction, consistent with subfunctionalization [104,105]. It has been proposed that certain paralogs may function as molecular buffers, supporting reproduction under chronic contaminant exposure but reducing plasticity in fluctuating environments [105]. This genomic diversification helps explain interspecific variation in VTG responses: for instance, D. rerio and medaka (Oryzias latipes) respond to ng/L estrogens, whereas salmonids typically require higher exposures [106]. From this perspective, genomic architecture provides a conceptual basis for interpreting interspecific differences in VTG induction and ecotoxicological sensitivity. Recent genomic analyses in Sichuan bream (Sinibrama taeniatus) identified six vtg genes, classified into types I–III based on subdomain structure, and confirmed that promoter sequences contain multiple estrogen response elements (EREs). The abundance of EREs correlated with the responsiveness of vtg expression to estrogen, with ERα emerging as the predominant driver of hepatic vtg transcription. Complementary to promoter-level regulation, recent work in the silver pomfret (Pampus argenteus) emphasized the importance of vitellogenin receptor (VGR) dynamics in shaping yolk accumulation. While vtg paralogs (vtgAa, vtgAb, vtgC) were primarily expressed in the liver, VGR expression in oocytes peaked during mid- to late vitellogenesis and localized to the plasma membrane, where it mediated VTG endocytosis [107]. This coordination between hepatic synthesis, plasma transport, and receptor availability in oocytes underscores that genomic diversification of vtg genes operates in tandem with the evolution of receptor-mediated uptake systems, ensuring efficient provisioning of yolk reserves during teleost reproduction.

3.3. Epigenetic Regulation of Sex-Specific Responses

Epigenetic mechanisms provide an additional layer of regulation, mediating both short- and long-term effects of EDCs. In D. rerio, the vtg1 promoter is hypermethylated in males and hypomethylated in females under baseline conditions. EE2 exposure reduces promoter methylation in males, enabling robust induction of vtg1 [108]. Genome-wide methylome analyses further show that EE2 alters methylation of steroidogenic loci, including cyp19a1a, star, and hsd17b [109]. Such changes can persist across generations. In O. latipes, ancestral exposure to BPA or EE2 led to reproductive defects in unexposed F2–F3 descendants, associated with stable germline methylation changes [110].
MicroRNAs also contribute. In D. rerio testes, EE2 activates the p53–miR-200 axis, altering the expression of genes involved in spermatogenesis and apoptosis, which in turn reduces sperm motility and compromises fertility [111]. Bisphenol S alters miRNAs targeting steroidogenic genes such as cyp19a1a, thereby interfering with estrogen signaling at the post-transcriptional level [112]. In M. galloprovincialis, BPA alters the expression of dnmt genes (DNA methyltransferases) [36], whereas in the oyster Crassostrea gigas, xenoestrogens disrupt non-coding RNAs and DNA methylation processes linked to gametogenesis [113]. Omics technologies expand this perspective by providing a system-level view of sex-specific responses. Transcriptomic studies in pipefish and salmonids show that EE2 upregulates female-biased genes such as vtg, zp2, and zp3 (zona pellucida proteins), and chg (choriogenins), together with regulators of lipid metabolism. Males often display stronger transcriptional shifts, likely reflecting their lower baseline estrogen activity [114].
Proteomic analyses corroborate these findings: in male sheepshead minnow (Cyprinodon variegatus), EE2 induced hepatic remodeling including VTG isoforms and metabolic enzymes [115]; in European flounder (P. flesus), field proteomics revealed co-induction of VTG, choriogenins, and HSP70/90 in polluted estuaries [77]. Metabolomics adds further evidence: in R. rutilus, EE2 decreased circulating androgens and estrogens in males, impairing fertility [33,116]. In Atlantic cod (G. morhua), combined RNA-Seq and metabolomics revealed hepatic reprogramming affecting immunity and lipid metabolism [117].
Evidence from invertebrates indicates comparable trends. In M. galloprovincialis, BPA exposure alters early embryogenesis [36], induces VTG-like proteins in males (sometimes associated with oxidative stress rather than purely estrogenic signaling) [118], and reprograms female gonadal metabolism under environmentally relevant concentrations [119]. Despite the limited availability of integrated omics studies, combined proteomic and metabolomic analyses with other contaminants (e.g., BDE-47, TBBPA) consistently demonstrate disruptions in energy metabolism, osmoregulation (particularly in females), and stress pathways. Comparable approaches in bivalves exposed to metals corroborate these responses [120].
VTG induction should not be regarded as an isolated endpoint but rather as part of a broader molecular fingerprint of estrogenic exposure. Its interpretation requires integration with other molecular and physiological pathways affected by EDCs [38,75,121,122]. While most evidence currently derives from biomedical and toxicological models, extending multi-omics to aquatic species, particularly when combined with machine learning, holds promise for identifying conserved signatures of estrogenic exposure and for distinguishing sex- and species-specific responses. These approaches could substantially advance predictive ecotoxicology [121,122].
Collectively, the different omics layers provide complementary insights into sex-specific responses to EDCs. Transcriptomics highlights large-scale transcriptional remodeling, including sex-dependent regulation of vtg and steroidogenic genes; proteomics validates these patterns at the protein level and reveals post-translational modifications; metabolomics connects molecular changes with altered energy balance, lipid metabolism, and reproductive performance; and epigenomics uncovers persistent modifications, such as DNA methylation and microRNA regulation, that may underlie transgenerational effects. Importantly, these approaches are not independent: when combined in multi-omics frameworks, they enable the identification of co-regulated pathways, position VTG within broader molecular networks, and strengthen causal inference for regulatory ecotoxicology. An overview of representative transcriptomic, proteomic, metabolomic, and epigenomic endpoints is provided in a dedicated table (see Table 5, Section 5). Complementing this overview, Table 2 provides a synthesis of key genes and regulatory elements in fish, mollusks, and crustaceans, highlighting their functions, modes of regulation, and sex-dependent responses.

4. Vitellogenin as a Biomarker

VTG is one of the most established and sensitive biomarkers of endocrine disruption in aquatic species. Normally expressed in oviparous females under estrogenic control, its ectopic induction in males and juveniles provides a robust indicator of estrogen receptor activation. In this section, VTG is examined from molecular and genomic perspectives, considering its structure, regulation, induction mechanisms, sex-specific differences, ecotoxicological applications, and limitations.

4.1. Structure, Function, and Gene Regulation of VTG

VTG is synthesized in the liver under estrogenic control and is normally expressed only in oviparous females during vitellogenesis. Its induction in males and juveniles is a hallmark of exogenous estrogenic exposure, making VTG a central biomarker in both laboratory assays and field biomonitoring. VTGs are large glycolipophosphoproteins that supply amino acids, lipids, and minerals to developing embryos. After hepatic synthesis, they are secreted into the bloodstream, transported to the ovary, endocytosed by oocytes, and cleaved into yolk proteins (lipovitellin, phosvitin, β′-components), each with distinct nutritive and regulatory functions. Comparative genomics has revealed diversification of vtg genes following genome duplication events. In D. rerio, the Aa/Ab clades are strongly estrogen-inducible, whereas the C clade (vtgC) is only weakly responsive [101,123]. CRISPR–Cas9 knockout studies confirmed that Aa/Ab paralogs are essential for oocyte maturation and fertility, while vtgC plays a secondary role [102]. In salmonids, which underwent an additional whole-genome duplication, more than ten paralogs have been identified, often arranged in chromosomal clusters, with marked differences in estrogen responsiveness [124]. Promoter analyses have highlighted multiple estrogen response elements (EREs) bound primarily by ERα (esr1), while ERβ isoforms (esr2a/b) contribute to transcriptional modulation [106]. Epigenetic regulation further refines expression: in D. rerio, the vtg1 promoter is hypermethylated in males and hypomethylated in females, and EE2 exposure reduces methylation in males, enabling strong induction [125]. Additional regulatory factors include HNF4α, a conserved hepatic transcription factor that regulates lipid and glucose metabolism and also contributes to the transcriptional control of vtg genes [126]. Among xenobiotic receptors, PXR is retained in several lineages and likely mediates crosstalk between VTG regulation, metabolism, and detoxification [127,128].

4.2. Mechanisms of Induction by EDCs and Receptor Crosstalk

Estrogens such as 17β-estradiol (E2) and synthetic analogs like EE2 activate ERα, which binds to vtg promoters and drives transcription. Robust VTG induction has been observed at environmentally relevant EE2 concentrations in D. rerio, O. latipes, and P. promelas [129,130]. Other xenoestrogens, including BPA and nonylphenol, also upregulate VTG, though with lower potency [131]. Crosstalk among signaling pathways modifies outcomes. Activation of AhR by dioxins or PCBs antagonizes ER signaling and suppresses VTG induction [97,132], whereas anti-androgens can enhance VTG expression [93]. Xenobiotic-sensing receptors, such as pregnane X receptor (PXR), reprogram estrogen-responsive networks and further modulate VTG regulation [119,120,121,122,123,124,125,126,127,128].
Comparable processes have been described in invertebrates. In M. galloprovincialis, nonylphenol exposure elevates VTG-like proteins in both sexes, commonly detected via alkali-labile phosphate assays [133]. BPA and related EDCs also induce VTG-like expression [15]. In crustaceans, organotins disrupt RXR-mediated pathways, interfering with metabolism and endocrine regulation [134]. RXR expression correlates with VTG during ovarian development, supporting its role in vitellogenesis [135].

4.3. Sex-Specific Differences in VTG Response

The diagnostic power of VTG stems from its sex-specific baseline. In females, VTG levels naturally rise during vitellogenesis and fluctuate with reproductive cycles, whereas in males and juveniles they remain negligible. This contrast makes contaminant-induced VTG expression a highly sensitive marker of estrogenic exposure. Laboratory studies consistently demonstrate stronger relative induction in males and juveniles, reflecting their low physiological background. Field observations corroborate this: in UK rivers impacted by wastewater effluents, male R. rutilus exhibited marked VTG elevation and intersex gonads, while females remained within physiological ranges. Juveniles also display heightened sensitivity, supporting their use as sentinel stages in ecotoxicological assays. Evidence from invertebrates indicates comparable trends, with VTG-like proteins detected in males following exposure to BPA, nonylphenol, or other xenoestrogens [19,34,136]. However, such responses can sometimes be confounded by generalized stress (e.g., oxidative stress), underscoring the need for careful interpretation. Overall, the sex-specific baseline of VTG remains one of its greatest strengths as a diagnostic endpoint, while variability in female physiology and potential non-estrogenic induction in invertebrates highlight its limitations.

4.4. Applications in Ecotoxicology

VTG is firmly established in ecotoxicological testing and regulatory frameworks, where it serves as a core endpoint in OECD Test Guidelines 229 (Fish Short-Term Reproduction Assay) and 234 (Fish Sexual Development Test) [86,87]. In these assays, VTG induction provides a sensitive readout of estrogenic activity and a mechanistic link between molecular signaling and reproductive outcomes. Several analytical methodologies are available. ELISA is the most widely applied method, providing high sensitivity and quantitative resolution, although it requires species-specific or cross-reactive antibodies that must be carefully validated. Western blotting offers semi-quantitative confirmation and is frequently used to verify ELISA results. At the transcript level, qPCR allows paralog-specific quantification of vtg genes, while RNA-Seq places VTG within broader co-expression networks, revealing links with genes involved in reproduction, metabolism, and stress responses. Non-invasive methods are emerging as innovative alternatives. Measurements of VTG in skin mucus or fin clips correlate strongly with plasma levels in several fish species, enabling more ethical, minimally invasive, and field-compatible monitoring strategies [136,137,138]. Biomonitoring studies consistently report elevated VTG levels and increased intersex prevalence in wild fish downstream of wastewater treatment plants, providing real-world evidence of estrogenic impacts. Within the AOP framework, ER activation constitutes the molecular initiating event, while vtg transcription and VTG protein synthesis represent early key events that connect molecular perturbation to impaired reproduction and altered population dynamics. VTG-like proteins are also being explored as biomarkers in bivalves and crustaceans, but their regulation remains poorly understood and their specificity as estrogenic indicators is debated [129,136]. Figure 1 integrates these methodological approaches with a schematic overview of the sex-specific induction of VTG by xenoestrogens in fish, highlighting detection through ELISA and Western blot. Despite its broad validation and regulatory use, the application of VTG as a biomarker is not without caveats. Interspecific variability, fluctuating baseline levels, and limitations in invertebrates highlight important challenges that need to be addressed, as discussed in the following section.

4.5. Limitations and Challenges

While VTG remains one of the most sensitive and widely validated biomarkers of estrogenic disruption, it is not suitable for evaluating the effects of several other classes of EDCs, including herbicides, anti-androgens, PFAS, organotins, and heavy metals, which act through distinct molecular pathways [122,139]. This underscores the importance of complementary biomarkers and multi-endpoint approaches to capture the full spectrum of endocrine disruption. Moreover, vtg is not expressed in the liver of most invertebrates, further limiting its use as a cross-phyla biomarker and highlighting the need for taxa-specific endpoints [19,37].
VTG is a robust and widely validated biomarker, but its application is not without challenges. Interspecific variation in vtg gene repertoires and differential estrogen-responsiveness of paralogs result in divergent inducibility profiles, complicating cross-species comparisons. In females, baseline fluctuations during reproductive cycles may obscure contaminant-induced responses, making interpretation highly context-dependent. Moreover, thresholds for ecological risk remain poorly defined: moderate VTG induction does not always predict reproductive impairment, indicating that induction levels cannot be linearly extrapolated to reproductive outcomes. Environmental mixtures add further complexity. AhR activation can antagonize ER-mediated VTG expression, whereas anti-androgens may potentiate it, and the net effect depends on interactions across multiple signaling pathways. Technical issues also persist, including antibody specificity, lack of standardized reference materials, integration of omics data, and inter-laboratory variability. Epigenetic regulation introduces an additional layer of complexity, since EE2 reduces vtg1 promoter methylation in D. rerio, while BPA alters DNA methylation and dnmt expression in M. galloprovincialis. These experimental findings illustrate how EDCs directly interfere with epigenetic machinery. More broadly, recent reviews emphasize that such molecular and epigenetic perturbations must be interpreted within wider mechanistic and ecological context [40,136].

4.6. Evolutionary and Biotechnological Perspectives of VTG

The diversification of vtg paralogs in teleosts and the occurrence of VTG-like proteins in mollusks and crustaceans reflect a conserved evolutionary strategy for yolk precursor synthesis under endocrine control. This variability underscores the ecogenomic significance of VTG: species with highly inducible paralogs act as sensitive sentinels of estrogenic pollution, whereas species with weakly inducible vtg genes may underestimate ecological risk. Beyond its ecological relevance, VTG also holds biotechnological potential. Advances in antibody development and multiplex immunoassays have improved diagnostic precision, while vtg promoters have been incorporated into biosensors and transgenic D. rerio reporters for real-time detection of estrogenic activity. Integrating these molecular tools with omics platforms and predictive models, including machine learning, could enhance assay sensitivity, enable species-specific customization, and expand monitoring capacities. Altogether, VTG remains a pivotal biomarker of endocrine disruption, but its greatest value emerges when interpreted in combination with complementary endpoints within multi-biomarker frameworks [79,107,138]. Table 3 summarizes vtg genes and paralogs across taxa, highlighting their physiological induction in females, induction under EDC exposure in males and juveniles, and the main regulatory pathways involved. The information is based on the studies presented in Section 4.

5. Integration with Other Biomarkers and Molecular Endpoints

Although VTG is a highly sensitive biomarker of estrogenic disruption, it reflects only part of the broader molecular and physiological networks affected by EDCs. Because real-world exposures typically involve mixtures acting through multiple pathways, integrating VTG with complementary endpoints enhances mechanistic specificity and ecological relevance.

5.1. Co-Expression with Hormone Metabolism and Stress Genes

In fish, VTG induction frequently coincides with the up-regulation of female-biased reproductive genes such as zp and chg, together with estrogen-responsive hepatic metabolic programs [140,141]. Conversely, activation of cyp1a by AhR agonists (e.g., dioxin-like PCBs) can antagonize ER-driven VTG responses, underscoring the importance of mixture effects and receptor cross-talk [98,140]. Stress proteins (HSP70/90) are also commonly detected alongside VTG in experimental and field studies. While non-specific, they strengthen WoE panels when interpreted in exposure context. For example, in mullet (Mugil cephalus), hepatic HSP70 and CYP1A reliably tracked contaminant exposure across estuaries [70,142].

5.2. Epigenetic and Transcriptomic Markers

Epigenetic endpoints extend interpretation beyond acute transcriptional changes. In adult male D. rerio, EE2 reduces CpG methylation at the vtg1 promoter, establishing a persistent hypomethylated state that facilitates robust re-induction upon re-exposure [143]. Across taxa, EDCs modulate DNA methylation machinery (e.g., dnmt genes) and non-coding RNAs (ncRNAs) involved in steroidogenesis and gametogenesis. Transcriptomic studies in flatfish and cyprinids consistently show that vtg induction co-occurs with zp and chg genes, together with immune and metabolic pathways, defining a broader “estrogenic signature” rather than an isolated endpoint [140,141]. In bivalves, BPA perturbs early embryonic transcription and produces sex-dependent metabolic shifts in gonads, reinforcing the value of combining VTG-like signals with multi-omics data [19,119].

5.3. Multi-Biomarker and AOP Approaches

Embedding VTG within WoE frameworks that integrate histological endpoints (e.g., intersex), receptor- and detoxification-pathway markers (e.g., cyp1a/CYP1A, HSPs), and omics data strengthens causal inference under complex exposure scenarios. Field proteomics in caged P. flesus revealed coordinated hepatic protein shifts consistent with mixed-contaminant stress, thereby complementing VTG-based diagnostics [144].
Within the AOP framework, ER activation constitutes the molecular initiating event (MIE), whereas increased vtg transcription and VTG protein synthesis represent early key events (KEs) that contribute to downstream gonadal pathology and reduced reproductive performance at organism and, ultimately, population levels [29,145].
Across taxa, EDCs also affect invertebrate biomarkers. In mollusks, alterations in DNA methyltransferases (DNMTs) and non-coding RNAs (ncRNAs) have been linked to disrupted gametogenesis [19,37], while in crustaceans, endocrine disruptors interfere with the EcR/RXR pathway and the methyl farnesoate (MF)–20-hydroxyecdysone (20E) axis, impairing molting and reproduction [83,84]. Table 4 summarizes key sex-specific biomarkers associated with EDCs in aquatic organisms, including their responses in males and females, detection methods, and ecotoxicological relevance.
Given the complexity of mixture exposures, integrative frameworks are increasingly required; the advantages and limitations of different omics strategies are outlined in Table 5.

6. Ecotoxicological and Ecogenomic Implications for Risk Assessment

The integration of molecular biomarkers such as VTG with complementary endpoints provides mechanistic insight into endocrine disruption, but a major challenge remains translating these signals into ecologically meaningful outcomes. In aquatic ecosystems, where organisms are chronically exposed to complex contaminant mixtures, risk assessment requires frameworks that link molecular events to adverse outcomes at population and community levels.

6.1. Species-Specific Sensitivity in Marine Fish

Marine fish display pronounced interspecific variability in their responses to endocrine disruptors. P. flesus consistently exhibits estrogenic fingerprints in impacted estuaries, with induction of VTG, choriogenins, and zona pellucida proteins in males, often accompanied by intersex development [150,151]. In M. cephalus, exposure to contaminated estuaries triggers broad transcriptional remodeling that integrates estrogenic, stress-related, and metabolic pathways, with co-induction of VTG, CYP1A, and HSP70 [70,152]. Recent studies also show that M. cephalus hepatocytes exposed to perfluorononanoic acid (PFNA) undergo estrogenic activation, with induction of vtg and estrogen receptor transcripts [153]. Seasonal and contaminant-driven shifts in liver and gonadal lipids further highlight metabolic involvement [154]. Field surveys confirm up-regulation of cyp19a1a and VTG, together with intersex in male M. cephalus, reinforcing their value as sentinel species [155]. In sea bass (Dicentrarchus labrax), responses are sex-specific: males exhibit robust VTG induction and epigenetic modifications, whereas females display subtler transcriptional shifts [156]. In G. morhua, EE2 exposure induces systemic hepatic reprogramming, altering immune and metabolic pathways with direct consequences for reproduction and health [157]. Collectively, these examples underline the importance of considering species-specific genomic and epigenomic repertoires in monitoring strategies.

6.2. Reproductive Fitness and Population-Level Effects

At the organismal level, molecular perturbations scale up to reproductive impairment. Long-term surveys of R. rutilus in UK rivers demonstrated clear links between feminization, intersex, impaired fertility, and exposure to estrogenic effluents [148]. A landmark whole-lake experiment in Canada revealed that chronic EE2 exposure caused P. promelas population collapse, with recovery only after removal of the contaminant source [149]. Comparable findings in P. flesus, M. cephalus, and G. morhua confirm that endocrine disruption impairs fecundity, sperm quality, oogenesis, and fertilization success, thereby bridging molecular biomarkers with demographic consequences [150,154,157].

6.3. Implications for Biodiversity and Ecosystem Health

The consequences of endocrine disruption extend beyond individuals to populations and ecosystems. Impaired reproduction in M. cephalus disrupts nutrient cycling and trophic transfer, while effects on commercially important species such as G. morhua and D. labrax raise concerns for fisheries sustainability.
In bivalves and oysters, altered vitellogenesis and gametogenesis compromise benthic–pelagic coupling and biofiltration. In crustaceans, interference with ecdysteroid-regulated molting and reproduction emphasizes ecosystem-level vulnerability [5,61]. While research on endocrine disruption in fish is extensive and has provided standardized biomarkers such as VTG, studies in mollusks and crustaceans are comparatively scarce and fragmented. This discrepancy highlights the urgent need for further investigations in non-fish taxa to broaden ecotoxicological paradigms. These findings underscore the importance of ecogenomic perspectives in predicting community shifts and biodiversity loss under contaminant pressure [8,158].

6.4. Integrating Molecular Data into Risk Assessment

Risk assessment frameworks are increasingly embedding molecular and organismal endpoints into regulatory contexts. Within the AOP framework, VTG induction is positioned as a key event linking ER activation to reproductive impairment and eventual population decline [159]. Advances in quantitative AOPs and networked AOPs are enhancing predictive capacity, whereas integrative strategies that combine biomarkers, histology, and chemical analyses strengthen causal inference.

7. Future Perspectives

Research on endocrine disruption in aquatic organisms has progressed from reliance on single biomarkers such as VTG toward integrated, multi-layered strategies that capture the complexity of sex- and species-specific responses. Nevertheless, significant knowledge gaps remain in developing diagnostic tools tailored to sex and developmental stage, exploiting multi-omics and big-data resources, and incorporating evolutionary and ecological dimensions. At the same time, embedding molecular endpoints into regulatory frameworks and sustainability agendas represents both a challenge and an opportunity.

7.1. Development of Sex-Specific Molecular Biomarkers

Although VTG induction in males and juveniles remains among the most sensitive indicators of estrogenic exposure, interpretation in females is complicated by natural reproductive cycles and high baseline levels. Future work should therefore prioritize sex-specific biomarker panels. Promising candidates include ER isoforms (esr1, esr2a, esr2b), oocyte-maturation genes (zp2, zp3), spermatogenesis markers (spag, sycp3), and sex-biased miRNAs regulating steroidogenesis and gametogenesis. Epigenetic signatures, such as sex-specific methylation patterns in vtg1 promoters or in steroidogenic regulators (cyp19a1a, hsd17b, star), also represent relatively stable biomarkers [147,160].
Establishing panels that discriminate male, female, and juvenile responses will increase diagnostic precision and reveal vulnerabilities masked when only VTG is measured. VTG will remain a cornerstone biomarker, but its diagnostic value can be enhanced by integrating omics signatures and improving immunoassay sensitivity and specificity. Recent advances include the development of species-specific assays in D. rerio and cyprinids [129,146], and customized antibodies against vitellogenin mRNA in crustaceans such as Palinurus elephas [161]. These strategies strengthen VTG-based diagnostics and support its central role in predictive and regulatory ecotoxicology.

7.2. Applications of Multi-Omics and Big Data

Multi-omics technologies now enable system-level profiling of endocrine disruption. Transcriptomic analyses in fish exposed to EE2 revealed genome-wide estrogenic signatures [114], while integrated transcriptomic–proteomic studies in D. rerio uncovered concurrent estrogenic and metabolic shifts [162]. In S. taeniatus, combined transcriptomic–metabolomic approaches identified key regulators of vitellogenesis [163]. Reviews highlight the growing use of triple-omics workflows to capture cross-taxa fingerprints [158], and proteomics of fish scales has identified estrogen-responsive markers for non-invasive diagnostics [164]. Transcriptomic approaches have identified toxicogenomic signatures, defined as conserved patterns of gene expression associated with specific contaminant classes or modes of action. These signatures further demonstrate the potential of omics integration to provide mechanistic insights and to distinguish between different exposure scenarios [165]. The next step is to translate these complex datasets into predictive tools. Machine learning and network analysis can integrate transcriptomic, proteomic, metabolomic, and epigenomic data to identify exposure signatures that are conserved yet sex-specific. Comparative ecogenomic databases will support cross-species predictions and help distinguish predominantly estrogenic exposures from mixed-contaminant profiles. These strategies will move ecotoxicology from descriptive diagnostics toward models capable of forecasting population-level outcomes.
Recent advances illustrate this potential. High-throughput transcriptomics (HTTr) in larval P. promelas generated transcriptomic points-of-departure (tPODs) that were comparable to—or more sensitive than—apical endpoints, facilitating rapid screening of contaminants such as PFAS [165,166,167,168]. Integrated multi-omics pipelines combined with machine learning are also being refined to improve robustness, interpretability, and regulatory acceptance [169,170,171].

7.3. Ecogenomics and Adaptive Evolution

An emerging focus concerns the evolutionary consequences of chronic exposure to EDCs. Variation in vtg paralogs, ER isoforms, and steroidogenic pathways across teleosts reflects both historical genome duplication and local adaptation. Increasing evidence suggests that populations chronically exposed to EDCs can undergo evolutionary shifts. Early studies mainly documented biochemical and epigenetic changes, such as altered steroidogenesis, promoter methylation, and microRNA regulation, which may persist across generations. In addition, population-level studies have demonstrated genetic adaptation to contaminant exposure. For example, killifish (Fundulus heteroclitus) populations inhabiting polluted estuaries have evolved resistance to dioxin-like compounds through adaptive variants in the aryl hydrocarbon receptor (AHR) pathway [172]. More broadly, evolutionary toxicology research has documented allelic shifts and genetic divergence in aquatic organisms chronically exposed to toxicants, confirming that long-term contaminant exposure can drive heritable evolutionary changes [173].
For example, R. rutilus populations in estrogen-contaminated UK rivers show altered baseline expression of vtg and ER genes [161,162], while M. galloprovincialis in polluted harbors display modified methylation profiles consistent with long-term selection [133]. Such adjustments may enhance survival under chronic stress but at the cost of reduced reproductive plasticity and resilience.
Future studies should employ genome-wide association studies (GWAS), population transcriptomics, and epigenetic-landscape analyses to clarify how adaptive responses shape biodiversity and persistence. Recent work also highlights the potential of DNA-methylation markers combined with machine learning as predictors of sex and stress history in fish [156,165,174]. Ecogenomics thus bridges molecular biomarkers and evolutionary ecology, showing how pollution not only impairs reproduction but can also drive long-term genetic and epigenetic change.
The stronger focus on fish compared to invertebrates reflects the current state of the literature, as molecular and omics studies in mollusks and crustaceans remain limited. Expanding research on these taxa is essential for a more comprehensive ecotoxicological perspective.

7.4. Regulatory and Sustainability Perspectives

Integrating molecular biomarkers into regulatory frameworks is increasingly recognized as a priority. The AOP framework formalizes links between MIE and population-level effects, and advances in quantitative and networked AOPs are strengthening predictive toxicology [159,175]. WoE approaches further enhance causal inference by combining molecular endpoints with histopathology and ecological data [176].
Future guidelines should incorporate omics-based signatures, epigenetic endpoints, and sex-specific panels to enhance sensitivity and predictive value. International initiatives, such as OECD recommendations, the EU Water Framework Directive, the Marine Strategy Framework Directive, and the US EPA’s EDSP program, already indicate regulatory uptake of molecular and genomic data [169,177,178]. Importantly, these efforts align with the United Nations SDG 14, underscoring how molecular ecotoxicology can provide both mechanistic insight and actionable strategies for biodiversity conservation and ecosystem management.

8. Conclusions

EDCs remain a critical environmental threat due to their ability to disrupt hormone-regulated pathways central to reproduction, development, and population stability in aquatic organisms. Among biomarkers of estrogenic disruption, VTG is the most sensitive and widely validated, providing a direct mechanistic link between estrogen receptor activation and reproductive impairment. Its diagnostic and predictive value is maximized when integrated into broader molecular, genomic, and physiological networks, and supported by epigenetic and multi-omics evidence. The ecogenomic perspective further demonstrates how differences in genomic architecture and regulatory repertoires shape species-specific sensitivity, with long-term studies such as those on R. rutilus in UK rivers illustrating the ecological benefits of reducing estrogenic effluents.
Looking forward, priorities include developing sex-specific biomarker panels, integrating multi-omics with machine learning, and expanding research on invertebrates to capture cross-phyla responses. Such integration will enhance ecological risk assessment and provide more robust tools for regulatory decision-making. Equally important is embedding molecular endpoints into regulatory frameworks, harmonizing protocols across regions, and aligning with international sustainability goals such as SDG 14.
In conclusion, VTG exemplifies how molecular biomarkers can transform ecotoxicology from a descriptive to a predictive science, providing actionable strategies for biodiversity conservation, ecosystem sustainability, and evidence-based environmental governance.

Author Contributions

Conceptualization, F.B.C., C.P., M.C.F. and A.P.; methodology, F.B.C. and A.P.; software, A.P.; validation, F.B.C., C.P., M.C.F. and A.P.; investigation, F.B.C., C.P., M.C.F. and A.P.; data curation, F.B.C. and A.P.; writing—original draft preparation, A.P.; writing—review and editing, F.B.C., C.P., M.C.F. and A.P.; visualization, F.B.C., C.P., M.C.F. and A.P.; supervision, A.P.; project administration, A.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ABCATP-Binding Cassette (transporters)
AOPAdverse Outcome Pathway
AOPsAdverse Outcome Pathways
ARAndrogen Receptor
ARαAndrogen Receptor alpha
ARβAndrogen Receptor beta
ATPAdenosine Triphosphate
AhRAryl Hydrocarbon Receptor
BDEBrominated Diphenyl Ether
BPABisphenol A
CRISPRClustered Regularly Interspaced Short Palindromic Repeats
CYP1ACytochrome P450 Family 1 Subfamily A
ChgChoriogenins
CpGCytosine-phosphate-Guanine dinucleotide
DDTDichlorodiphenyltrichloroethane
DNADeoxyribonucleic Acid
E217β-Estradiol
EDCEndocrine-Disrupting Chemical
EDCsEndocrine-Disrupting Chemicals
EDSPEndocrine Disruptor Screening Program (US EPA)
EE217α-Ethinylestradiol
ELISAEnzyme-Linked Immunosorbent Assay
EREstrogen Receptor
EREEstrogen Response Element
ERαEstrogen Receptor alpha
ERβEstrogen Receptor beta
esr2aEstrogen Receptor beta a (teleost isoform)
esr2bEstrogen Receptor beta b (teleost isoform)
EUEuropean Union
GPERG Protein-Coupled Estrogen Receptor
GWASGenome-Wide Association Study
HNF4αHepatocyte Nuclear Factor 4 alpha
HSP70Heat Shock Protein 70
HSP90Heat Shock Protein 90
HSPsHeat Shock Proteins
HTTrHigh-Throughput Transcriptomics
MIEMolecular Initiating Event (AOP framework)
OECDOrganisation for Economic Co-operation and Development
PCBPolychlorinated Biphenyl
PFASPer- and Polyfluoroalkyl Substances
PFNAPerfluorononanoic Acid
PXRPregnane X Receptor
RNARibonucleic Acid
RNA-SeqRNA Sequencing
RXRRetinoid X Receptor
SDGSustainable Development Goal
SLCSolute Carrier Transporter
StARSteroidogenic Acute Regulatory Protein
tPODTranscriptomic Point of Departure
VTGVitellogenin
WoEWeight of Evidence
ZP2Zona Pellucida Glycoprotein 2
ZP3Zona Pellucida Glycoprotein 3
cyp19a1aCytochrome P450 Family 19 Subfamily A Member 1a (ovarian aromatase)
cyp19a1bCytochrome P450 Family 19 Subfamily A Member 1b (brain aromatase)
dnmtDNA Methyltransferase
mPRαMembrane Progestin Receptor alpha
miR-200MicroRNA-200 family
ncRNANon-Coding RNA
qPCRQuantitative Polymerase Chain Reaction

References

  1. Wang, Y.; Zhou, J. Endocrine disrupting chemicals in aquatic environments: A potential reason for organism extinction? Aquat. Ecosyst. Health Manag. 2013, 16, 88–93. [Google Scholar] [CrossRef]
  2. Sumpter, J.P.; Johnson, A.C. Lessons from endocrine disruption and their application to other issues concerning trace organics in the aquatic environment. Environ. Sci. Technol. 2005, 39, 4321–4332. [Google Scholar] [CrossRef] [PubMed]
  3. Vulliet, E.; Cren-Olivé, C. Screening of pharmaceuticals and hormones at the regional scale, in surface and groundwaters intended to human consumption. Environ. Pollut. 2011, 159, 2929–2934. [Google Scholar] [CrossRef] [PubMed]
  4. Kolpin, D.W.; Furlong, E.T.; Meyer, M.T.; Thurman, E.M.; Zaugg, S.D.; Barber, L.B.; Buxton, H.T. Pharmaceuticals, hormones, and other organic wastewater contaminants in U.S. streams, 1999–2000: A national reconnaissance. Environ. Sci. Technol. 2002, 36, 1202–1211. [Google Scholar] [CrossRef]
  5. Beyer, J.; Green, N.W.; Brooks, S.; Allan, I.J.; Ruus, A.; Gomes, T.; Bråte, I.L.N.; Schøyen, M. Blue mussels (Mytilus edulis spp.) as sentinel organisms in coastal pollution monitoring: A review. Mar. Environ. Res. 2017, 130, 338–365. [Google Scholar] [CrossRef]
  6. Diamanti-Kandarakis, E.; Bourguignon, J.P.; Giudice, L.C.; Hauser, R.; Prins, G.S.; Soto, A.M.; Zoeller, R.T.; Gore, A.C. Endocrine-disrupting chemicals: An Endocrine Society scientific statement. Endocr. Rev. 2009, 30, 293–342. [Google Scholar] [CrossRef]
  7. Martyniuk, C.J.; Martínez, R.; Navarro-Martín, L.; Kamstra, J.H.; Schwendt, A.; Reynaud, S.; Chalifour, L. Emerging concepts and opportunities for endocrine disruptor screening of the non-EATS modalities. Environ. Res. 2022, 204 Pt A, 111904. [Google Scholar] [CrossRef]
  8. Windsor, F.M.; Ormerod, S.J.; Tyler, C.R. Endocrine disruption in aquatic systems: Up-scaling research to address ecological consequences. Biol. Rev. 2018, 93, 626–641. [Google Scholar] [CrossRef]
  9. Crane, M.; Hallmark, N.; Lagadic, L.; Ott, K.; Pickford, D.; Preuss, T.; Thompson, H.; Thorbek, P.; Weltje, L.; Wheeler, J.R. Establishing the relevance of endocrine-disrupting effects for nontarget vertebrate populations. Integr. Environ. Assess. Manag. 2019, 15, 299–300. [Google Scholar] [CrossRef]
  10. Major, K.M.; DeCourten, B.M.; Li, J.; Britton, M.; Settles, M.L.; Mehinto, A.C.; Connon, R.E.; Brander, S.M. Early-life exposure to environmentally relevant levels of endocrine disruptors drive multigenerational and transgenerational epigenetic changes in a fish model. Front. Mar. Sci. 2020, 7, 471. [Google Scholar] [CrossRef]
  11. Ayobahan, S.U.; Eilebrecht, E.; Kotthoff, M.; Baumann, L.; Eilebrecht, S.; Teigeler, M.; Hollert, H.; Kalkhof, S.; Schäfers, C. A combined FSTRA-shotgun proteomics approach to identify molecular changes in zebrafish upon chemical exposure. Sci. Rep. 2019, 9, 6599. [Google Scholar] [CrossRef] [PubMed]
  12. Celino-Brady, F.T.; Lerner, D.T.; Seale, A.P. Experimental approaches for characterizing the endocrine-disrupting effects of environmental chemicals in fish. Front. Endocrinol. 2021, 11, 619361. [Google Scholar] [CrossRef]
  13. Barra, R.O.; Chiang, G.; Saavedra, M.F.; Orrego, R.; Servos, M.R.; Hewitt, L.M.; McMaster, M.E.; Bahamonde, P.; Tucca, F.; Munkittrick, K.R.; et al. Endocrine disruptor impacts on fish from Chile: The influence of wastewaters. Front. Endocrinol. 2021, 12, 611281. [Google Scholar] [CrossRef]
  14. Tseng, P.W.; Wu, G.C.; Kuo, W.L.; Tseng, Y.C.; Chang, C.F. The ovarian transcriptome at the early stage of testis removal-induced male-to-female sex change in the protandrous black porgy Acanthopagrus schlegelii. Front. Genet. 2022, 13, 816955. [Google Scholar] [CrossRef]
  15. Matozzo, V.; Gagné, F.; Marin, M.G.; Ricciardi, F.; Blaise, C. Vitellogenin as a biomarker of exposure to estrogenic compounds in aquatic invertebrates: A review. Environ. Int. 2008, 34, 531–545. [Google Scholar] [CrossRef]
  16. Hutchinson, T.H.; Ankley, G.T.; Segner, H.; Tyler, C.R. Screening and testing for endocrine disruption in fish—Biomarkers as “signposts,” not “traffic lights,” in risk assessment. Environ. Health Perspect. 2006, 114 (Suppl. S1), 106–114. [Google Scholar] [CrossRef]
  17. Bowman, C.J.; Kroll, K.J.; Hemmer, M.J.; Folmar, L.C.; Denslow, N.D. Estrogen-Induced Vitellogenin mRNA and Protein in Sheepshead Minnow (Cyprinodon variegatus). Gen. Comp. Endocrinol. 2000, 120, 300–313. [Google Scholar] [CrossRef]
  18. Brown, R.J.; Panter, G.H.; Burden, N.; Salinas, E.R.; Weltje, L.; Wheeler, J.R.; Wolf, Y.; Lagadic, L. Are changes in vitellogenin concentrations in fish reliable indicators of chemical-induced endocrine activity? Ecotoxicol. Environ. Saf. 2023, 266, 115563. [Google Scholar] [CrossRef] [PubMed]
  19. Tran, T.K.A.; Yu, R.M.K.; Islam, R.; Nguyen, T.H.T.; Bui, T.L.H.; Kong, R.Y.C.; O’Connor, W.A.; Leusch, F.D.L.; Andrew-Priestley, M.; MacFarlane, G.R. The utility of vitellogenin as a biomarker of estrogenic endocrine disrupting chemicals in molluscs. Environ. Pollut. 2019, 248, 1067–1078. [Google Scholar] [CrossRef]
  20. Ketata, I.; Denier, X.; Hamza-Chaffai, A.; Minier, C. Endocrine-related reproductive effects in molluscs. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2008, 147, 261–270. [Google Scholar] [CrossRef] [PubMed]
  21. Rodríguez, E.M.; Medesani, D.A.; Fingerman, M. Endocrine disruption in crustaceans due to pollutants: A review. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2007, 146, 661–671. [Google Scholar] [CrossRef] [PubMed]
  22. Palmer, B.D.; Huth, L.K.; Pieto, D.L.; Selcer, K.W. Vitellogenin as a biomarker for xenobiotic estrogens in an amphibian model system. Environ. Toxicol. Chem. 1998, 17, 30–36. [Google Scholar] [CrossRef]
  23. Tompsett, A.R.; Wiseman, S.; Higley, E.; Pryce, S.; Chang, H.; Giesy, J.P.; Hecker, M. Effects of 17α-ethynylestradiol on sexual differentiation and development of the African clawed frog (Xenopus laevis). Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2012, 156, 202–210. [Google Scholar] [CrossRef] [PubMed]
  24. Santos, E.M.; Ball, J.S.; Williams, T.D.; Wu, H.; Ortega, F.; Van Aerle, R.; Katsiadaki, I.; Falciani, F.; Viant, M.R.; Chipman, J.K.; et al. Identifying health impacts of exposure to copper using transcriptomics and metabolomics in a fish model. Environ. Sci. Technol. 2010, 44, 820–826. [Google Scholar] [CrossRef]
  25. Viant, M.R.; Rosenblum, E.S.; Tjeerdema, R.S. NMR-based metabolomics: A powerful approach for characterizing the effects of environmental stressors on organism health. Environ. Sci. Technol. 2003, 37, 4982–4989. [Google Scholar] [CrossRef]
  26. Marjan, P.; Martyniuk, C.J.; Arlos, M.J.; Servos, M.R.; Ruecker, N.J.; Munkittrick, K.R. Identifying transcriptomic indicators of tertiary treated municipal effluent in longnose dace (Rhinichthys cataractae) caged under semi-controlled conditions in experimental raceways. Sci. Total Environ. 2024, 923, 171257. [Google Scholar] [CrossRef]
  27. United Nations. Sustainable Development Goal 14: Life Below Water. Available online: https://sdgs.un.org/goals/goal14 (accessed on 24 August 2025).
  28. Kloas, W.; Urbatzka, R.; Opitz, R.; Würtz, S.; Behrends, T.; Hermelink, B.; Hofmann, F.; Jagnytsch, O.; Kroupova, H.; Lorenz, C.; et al. Endocrine disruption in aquatic vertebrates. Ann. N.Y. Acad. Sci. 2009, 1163, 187–200. [Google Scholar] [CrossRef]
  29. Ankley, G.T.; Bencic, D.C.; Breen, M.S.; Collette, T.W.; Conolly, R.B.; Denslow, N.D.; Edwards, S.W.; Ekman, D.R.; Garcia-Reyero, N.; LaLone, C.A.; et al. Endocrine disrupting chemicals in fish: Developing exposure indicators and predictive models of effects based on mechanism of action. Aquat. Toxicol. 2009, 92, 168–178. [Google Scholar] [CrossRef]
  30. Porte, C.; Janer, G.; Lorusso, L.C.; Ortiz-Zarragoitia, M.; Cajaraville, M.P.; Fossi, M.C.; Canesi, L. Endocrine disruptors in marine organisms: Approaches and perspectives. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2006, 143, 303–315. [Google Scholar] [CrossRef]
  31. Tasselli, S.; Marziali, L.; Roscioli, C.; Guzzella, L. Legacy Dichlorodiphenyltrichloroethane (DDT) Pollution in a River Ecosystem: Sediment Contamination and Bioaccumulation in Benthic Invertebrates. Sustainability 2023, 15, 6493. [Google Scholar] [CrossRef]
  32. Hayes, T.B.; Anderson, L.L.; Beasley, V.R.; de Solla, S.R.; Iguchi, T.; Ingraham, H.; Kestemont, P.; Kniewald, J.; Kniewald, Z.; Langlois, V.S.; et al. Demasculinization and feminization of male gonads by atrazine: Consistent effects across vertebrate classes. J. Steroid Biochem. Mol. Biol. 2011, 127, 64–73. [Google Scholar] [CrossRef] [PubMed]
  33. de Lima Oliveira, W.; Mendonça Mota, T.F.; da Silva, A.P.; de Lima Oliveira, R.D.; Comelli, C.L.; Orlandini, N.D.; Zimmer, D.F.; de Oliveira, E.C.; de Castilhos Ghisi, N. Does the atrazine increase animal mortality: Unraveling through a meta-analytic study. Sci. Total Environ. 2024, 951, 175553. [Google Scholar] [CrossRef] [PubMed]
  34. Yuan, M.; Chen, S.; Zeng, C.; Fan, Y.; Ge, W.; Chen, W. Estrogenic and non-estrogenic effects of bisphenol A and its action mechanism in the zebrafish model: An overview of the past two decades of work. Environ. Int. 2023, 176, 107976. [Google Scholar] [CrossRef] [PubMed]
  35. Tyler, C.R.; Lange, A.; Paull, G.C.; Katsu, Y.; Iguchi, T. The roach (Rutilus rutilus) as a sentinel for assessing endocrine disruption. Environ. Sci. 2007, 14, 235–253. [Google Scholar] [CrossRef]
  36. Balbi, T.; Franzellitti, S.; Fabbri, R.; Montagna, M.; Fabbri, E.; Canesi, L. Impact of bisphenol A (BPA) on early embryo development in the marine mussel Mytilus galloprovincialis: Effects on gene transcription. Environ. Pollut. 2016, 218, 996–1004. [Google Scholar] [CrossRef]
  37. Fernández-González, L.E.; Sánchez-Marín, P.; Gestal, C.; Beiras, R.; Diz, A.P. Vitellogenin gene expression in marine mussels exposed to ethinylestradiol: No induction at the transcriptional level. Mar. Environ. Res. 2021, 168, 105315. [Google Scholar] [CrossRef]
  38. Kidd, K.A.; Blanchfield, P.J.; Mills, K.H.; Palace, V.P.; Evans, R.E.; Lazorchak, J.M.; Flick, R.W. Collapse of a fish population after exposure to a synthetic estrogen. Proc. Natl. Acad. Sci. USA 2007, 104, 8897–8901. [Google Scholar] [CrossRef]
  39. Blanchfield, P.J.; Kidd, K.A.; Docker, M.F.; Palace, V.P.; Park, B.J.; Postma, L.D. Recovery of a wild fish population from whole-lake additions of a synthetic estrogen. Environ. Sci. Technol. 2015, 49, 3136–3144. [Google Scholar] [CrossRef]
  40. Marlatt, V.L.; Bayen, S.; Castaneda-Cortès, D.; Delbès, G.; Grigorova, P.; Langlois, V.S.; Martyniuk, C.J.; Metcalfe, C.D.; Parent, L.; Rwigemera, A.; et al. Impacts of endocrine disrupting chemicals on reproduction in wildlife and humans. Environ. Res. 2022, 208, 112584. [Google Scholar] [CrossRef]
  41. Hahn, M.E. Aryl hydrocarbon receptors: Diversity and evolution. Chem. Biol. Interact. 2002, 141, 131–160. [Google Scholar] [CrossRef]
  42. Cai, Z.; Zhou, G.; Yu, X.; Du, Y.; Man, Q.; Wang, W.C. Perfluorooctanoic acid disrupts thyroid hormone biosynthesis by altering glycosylation of Na+/I− symporter in larval zebrafish. Ecotoxicol. Environ. Saf. 2025, 297, 118249. [Google Scholar] [CrossRef]
  43. Yu, Q.; Cai, L.; Liu, H.; Hu, Z.; Wu, H.; Chen, X.; Ren, C.; Zhang, J. Bioaccumulation and bioelimination of per- and polyfluoroalkyl substances in aquatic organisms: A review. J. Water Process Eng. 2025, 75, 108065. [Google Scholar] [CrossRef]
  44. Fabrello, J.; Targhetta, F.; Ciscato, M.; Asnicar, D.; Bernardini, I.; Milan, M.; Patarnello, T.; Marin, M.G.; Matozzo, V. First Evidence of In Vitro Effects of C6O4—A Substitute of PFOA—On Haemocytes of the Clam Ruditapes philippinarum. Toxics 2021, 9, 191. [Google Scholar] [CrossRef] [PubMed]
  45. Aquilina-Beck, A.A.; Reiner, J.L.; Chung, K.W.; DeLise, M.J.; Key, P.B.; DeLorenzo, M.E. Uptake and Biological Effects of Perfluorooctane Sulfonate Exposure in the Adult Eastern Oyster Crassostrea virginica. Arch. Environ. Contam. Toxicol. 2020, 79, 333–342. [Google Scholar] [CrossRef] [PubMed]
  46. Romersi, R.F.; Nicklisch, S.C.T. Interactions of Environmental Chemicals and Natural Products with ABC and SLC Transporters in the Digestive System of Aquatic Organisms. Front. Physiol. 2022, 12, 767766. [Google Scholar] [CrossRef]
  47. Bieczynski, F.; Painefilú, J.C.; Venturino, A.; Luquet, C.M. Expression and Function of ABC Proteins in Fish Intestine. Front. Physiol. 2021, 12, 791834. [Google Scholar] [CrossRef]
  48. Ervin, S.M.; Li, H.; Lim, L.; Roberts, L.R.; Liang, X.; Mani, S.; Redinbo, M.R. Gut Microbial β-Glucuronidases Reactivate Estrogens as Components of the Estrobolome. J. Biol. Chem. 2019, 294, 18586–18599. [Google Scholar] [CrossRef]
  49. Ma, L.; Yates, S.R. Degradation and Metabolite Formation of 17β-Estradiol-3-Glucuronide and 17β-Estradiol-3-Sulphate in River Water and Sediment. Water Res. 2018, 139, 1–9. [Google Scholar] [CrossRef]
  50. Monteverdi, G.H.; Di Giulio, R.T. In Vitro and In Vivo Association of 2,3,7,8-Tetrachlorodibenzo-p-dioxin and Benzo[a]pyrene with the Yolk-Precursor Protein Vitellogenin. Environ. Toxicol. Chem. 2000, 19, 2502–2511. [Google Scholar] [CrossRef]
  51. Heiden, T.K.; Hutz, R.J.; Carvan, M.J. Accumulation, Tissue Distribution, and Maternal Transfer of Dietary 2,3,7,8-Tetrachlorodibenzo-p-dioxin: Impacts on Reproductive Success of Zebrafish. Toxicol. Sci. 2005, 87, 497–507. [Google Scholar] [CrossRef]
  52. Honda, M.; Muta, A.; Akasaka, T.; Inoue, Y.; Shimasaki, Y.; Kannan, K.; Okino, N.; Oshima, Y. Identification of Perfluorooctane Sulfonate Binding Protein in the Plasma of Tiger Pufferfish Takifugu rubripes. Ecotoxicol. Environ. Saf. 2014, 104, 409–413. [Google Scholar] [CrossRef]
  53. Point, A.D.; Crimmins, B.S.; Holsen, T.M.; Fernando, S.; Hopke, P.K.; Darie, C.C. Can Blood Proteome Diversity among Fish Species Help Explain Perfluoroalkyl Acid Trophodynamics in Aquatic Food Webs? Sci. Total Environ. 2023, 875, 162337. [Google Scholar] [CrossRef]
  54. Molnár, S.; Kulcsár, G.; Perjési, P. Determination of Steroid Hormones in Water Samples by Liquid Chromatography Electrospray Ionization Mass Spectrometry Using Parallel Reaction Monitoring. Microchem. J. 2022, 175, 107105. [Google Scholar] [CrossRef]
  55. Shao, Z.; Guagliardo, P.; Jiang, H.; Wang, W.-X. Intra- and Intercellular Silver Nanoparticle Translocation and Transformation in Oyster Gill Filaments: Coupling Nanoscale SIMS and Dual Stable Isotope Tracing. Environ. Sci. Technol. 2021, 55, 433–446. [Google Scholar] [CrossRef]
  56. Ikuta, T.; Tame, A.; Takahashi, T.; Nomaki, H.; Nakajima, R. Microplastic Particles Are Phagocytosed in Gill Cells of Deep-Sea and Coastal Mussels. Front. Mar. Sci. 2022, 9, 1034950. [Google Scholar] [CrossRef]
  57. Dong, L.; Sun, Y.; Chu, M.; Xie, Y.; Wang, P.; Li, B.; Li, Z.; Xu, X.; Feng, Y.; Sun, G.; et al. Exploration of Response Mechanisms in the Gills of Pacific Oyster (Crassostrea gigas) to Cadmium Exposure through Integrative Metabolomic and Transcriptomic Analyses. Animals 2024, 14, 2318. [Google Scholar] [CrossRef] [PubMed]
  58. Martínez-Escauriaza, R.; Lozano, V.; Pérez-Parallé, M.L.; Blanco, J.; Sánchez, J.L.; Pazos, A.J. Expression Analyses of Genes Related to Multixenobiotic Resistance in Mytilus galloprovincialis after Exposure to Dinophysis acuminata. Toxins 2021, 13, 614. [Google Scholar] [CrossRef] [PubMed]
  59. Qin, K.; Wu, L.; Fu, S.; Que, H.; Shi, B. Transcriptomic Analysis Reveals the Mechanisms of Cadmium Transport and Detoxification in Portuguese Oysters (Crassostrea angulata). Animals 2025, 15, 1041. [Google Scholar] [CrossRef]
  60. Banaee, M.; Zeidi, A.; Mikušková, N.; Faggio, C. Assessing Metal Toxicity on Crustaceans in Aquatic Ecosystems: A Comprehensive Review. Biol. Trace Elem. Res. 2024, 202, 5743–5761. [Google Scholar] [CrossRef]
  61. Chen, Y.; Sun, X.; Du, J.; Hu, J.; Bao, Z.; Qu, Z. Effects of Methyl Farnesoate on the Growth and Antioxidant Capacity of Neocaridina denticulata. Antioxidants 2025, 14, 635. [Google Scholar] [CrossRef]
  62. Rodríguez, E.M. Endocrine Disruption in Crustaceans: New Findings and Perspectives. Mol. Cell. Endocrinol. 2024, 585, 112189. [Google Scholar] [CrossRef]
  63. Jones, S.E.; Gutkowski, N.; Demick, S.; Curello, M.; Pavia, A.; Robuck, A.R.; Li, M.-L. Assessing Bivalves as Biomonitors of Per- and Polyfluoroalkyl Substances in Coastal Environments. Environ. Sci. Technol. 2025, 59, 5202–5213. [Google Scholar] [CrossRef] [PubMed]
  64. Oikari, A. Use of early juvenile zebrafish (Danio rerio) for in vivo assessment of endocrine modulation by xenoestrogens. J. Environ. Anal. Toxicol. 2013, 4, 202. [Google Scholar] [CrossRef]
  65. Piferrer, F.; Anastasiadi, D.; Valdivieso, A.; Sánchez-Baizán, N.; Moraleda-Prados, J.; Ribas, L. The model of the conserved epigenetic regulation of sex. Front. Genet. 2019, 10, 857. [Google Scholar] [CrossRef]
  66. Dang, Z.C. Fish biomarkers for regulatory identification of endocrine disrupting chemicals. Environ. Pollut. 2014, 185, 266–270. [Google Scholar] [CrossRef] [PubMed]
  67. Wang, Y.Y.; Sun, L.X.; Zhu, J.J.; Zhao, Y.; Wang, H.; Liu, H.J.; Ji, X.S. Epigenetic control of cyp19a1a expression is critical for high temperature induced Nile tilapia masculinization. J. Therm. Biol. 2017, 69, 76–84. [Google Scholar] [CrossRef]
  68. Thomas, P. Membrane Progesterone Receptors (mPRs, PAQRs): Review of Structural and Signaling Characteristics. Cells 2022, 11, 1785. [Google Scholar] [CrossRef]
  69. Nagata, J.; Mushirobira, Y.; Nishimiya, O.; Yamaguchi, Y.; Fujita, T.; Hiramatsu, N.; Hara, A.; Todo, T. Hepatic estrogen-responsive genes relating to oogenesis in cutthroat trout (Oncorhynchus clarki): Transcriptional induction in primary cultured hepatocytes and in vitro promoter transactivation in response to estradiol-17β. Gen. Comp. Endocrinol. 2021, 310, 113812. [Google Scholar] [CrossRef]
  70. Maradonna, F.; Carnevali, O. Vitellogenin, zona radiata protein, cathepsin D and heat shock protein 70 as biomarkers of exposure to xenobiotics. Biomarkers 2007, 12, 240–255. [Google Scholar] [CrossRef]
  71. Canesi, L.; Betti, M.; Lorusso, L.C.; Ciacci, C.; Gallo, G. In vivo effects of bisphenol A in Mytilus hemocytes: Modulation of kinase-mediated signalling pathways. Aquat. Toxicol. 2005, 71, 73–84. [Google Scholar] [CrossRef]
  72. Qu, M.; Xu, J.; Yang, Y.; Li, R.; Li, T.; Chen, S.; Di, Y. Assessment of sulfamethoxazole toxicity to marine mussels (Mytilus galloprovincialis): Combining p38-MAPK signaling pathway modulation with histopathological alterations. Ecotoxicol. Environ. Saf. 2023, 249, 114365. [Google Scholar] [CrossRef]
  73. Kundu, S.; Ray, A.; Das Gupta, S.; Biswas, A.; Roy, S.; Tiwari, N.K.; Kumar, V.S.; Das, B.K. Environmental bisphenol A disrupts methylation of steroidogenic genes in the ovary of paradise threadfin (Polynemus paradiseus) via abnormal DNA methylation: Implications for human exposure and health risk assessment. Chemosphere 2024, 351, 141236. [Google Scholar] [CrossRef] [PubMed]
  74. Breves, J.P.; Duffy, T.A.; Einarsdottir, I.E.; Björnsson, B.T.; McCormick, S.D. In vivo effects of 17α-ethinylestradiol, 17β-estradiol and 4-nonylphenol on insulin-like growth factor-binding proteins (IGFBPs) in Atlantic salmon. Aquat. Toxicol. 2018, 203, 28–39. [Google Scholar] [CrossRef] [PubMed]
  75. Jobling, S.; Williams, R.; Johnson, A.; Taylor, A.; Gross-Sorokin, M.; Nolan, M.; Tyler, C.R.; van Aerle, R.; Santos, E.; Brighty, G. Predicted exposures to steroid estrogens in U.K. rivers correlate with widespread sexual disruption in wild fish populations. Environ. Health Perspect. 2006, 114 (Suppl. S1), 32–39. [Google Scholar] [CrossRef] [PubMed]
  76. Aluru, N.; Hallanger, I.G.; McMonagle, H.; Harju, M. Hepatic gene expression profiling of Atlantic cod (Gadus morhua) liver after exposure to organophosphate flame retardants revealed altered cholesterol biosynthesis and lipid metabolism. Environ. Toxicol. Chem. 2021, 40, 1639–1648. [Google Scholar] [CrossRef]
  77. Falciani, F.; Diab, A.M.; Sabine, V.; Williams, T.; Ortega, F.; George, S.G.; Chipman, J. Hepatic transcriptomic profiles of European flounder (Platichthys flesus) from field sites and computational approaches to predict site from stress gene responses following exposure to model toxicants. Aquat. Toxicol. 2008, 90, 92–101. [Google Scholar] [CrossRef]
  78. Blalock, B.; Robinson, W.E.; Loguinov, A.; Vulpe, C.D.; Krick, K.S.; Poynton, H.C. Transcriptomic and network analyses reveal mechanistic-based biomarkers of endocrine disruption in the marine mussel, Mytilus edulis. Environ. Sci. Technol. 2018, 52, 9419–9430. [Google Scholar] [CrossRef]
  79. Jubeaux, G.; Audouard-Combe, F.; Simon, R.; Tutundjian, R.; Salvador, A.; Geffard, O.; Chaumot, A. Vitellogenin-like proteins among invertebrate species diversity: Potential of proteomic mass spectrometry for biomarker development. Environ. Sci. Technol. 2012, 46, 6315–6323. [Google Scholar] [CrossRef]
  80. Breitholtz, M. Crustaceans. In Endocrine Disrupters; John Wiley & Sons, Ltd.: Chichester, UK, 2013; pp. 116–142. [Google Scholar] [CrossRef]
  81. Vogt, É.L.; Model, J.F.A.; Vinagre, A.S. Effects of organotins on crustaceans: Update and perspectives. Front. Endocrinol. 2018, 9, 65. [Google Scholar] [CrossRef]
  82. Mazurová, E.; Hilscherová, K.; Triebskorn, R.; Köhler, H.R. Endocrine regulation of reproduction in crustaceans: Identification of potential targets for toxicants and environmental contaminants. Biologia 2008, 63, 139–150. [Google Scholar] [CrossRef]
  83. LeBlanc, G.A. Crustacean endocrine toxicology: A review. Ecotoxicology 2007, 16, 61–81. [Google Scholar] [CrossRef]
  84. Toyota, K.; Yamane, F.; Ohira, T. Impacts of Methyl Farnesoate and 20-Hydroxyecdysone on Larval Mortality and Metamorphosis in the Kuruma Prawn Marsupenaeus japonicus. Front. Endocrinol. 2020, 11, 475. [Google Scholar] [CrossRef]
  85. Knigge, T.; LeBlanc, G.A.; Ford, A.T. A crab is not a fish: Unique aspects of the crustacean endocrine system and considerations for endocrine toxicology. Front. Endocrinol. 2021, 12, 587608. [Google Scholar] [CrossRef] [PubMed]
  86. OECD. Test No. 229: Fish Short Term Reproduction Assay; OECD Guidelines for the Testing of Chemicals, Section 2; OECD Publishing: Paris, France, 2012. [Google Scholar] [CrossRef]
  87. OECD. Test No. 234: Fish Sexual Development Test; OECD Guidelines for the Testing of Chemicals, Section 2; OECD Publishing: Paris, France, 2011. [Google Scholar] [CrossRef]
  88. Filby, A.L.; Tyler, C.R. Molecular characterization of estrogen receptors 1, 2a, and 2b and their tissue and ontogenic expression profiles in fathead minnow (Pimephales promelas). Biol. Reprod. 2005, 73, 648–662. [Google Scholar] [CrossRef] [PubMed]
  89. Bardet, P.; Horard, B.; Robinson-Rechavi, M.; Laudet, V.; Vanacker, J. Characterization of oestrogen receptors in zebrafish (Danio rerio). J. Mol. Endocrinol. 2002, 28, 153–163. [Google Scholar] [CrossRef]
  90. Griffin, L.B.; January, K.E.; Ho, K.W.; Cotter, K.A.; Callard, G.V. Morpholino-mediated knockdown of ERα, ERβa, and ERβb mRNAs in zebrafish (Danio rerio) embryos reveals differential regulation of estrogen-inducible genes. Endocrinology 2013, 154, 4158–4169. [Google Scholar] [CrossRef] [PubMed]
  91. Pinto, P.I.; Estêvão, M.D.; Power, D.M. Effects of estrogens and estrogenic disrupting compounds on fish mineralized tissues. Mar. Drugs 2014, 12, 4474–4494. [Google Scholar] [CrossRef]
  92. Ryu, T.; Okamoto, K.; Ansai, S.; Nakao, M.; Kumar, A.; Iguchi, T.; Ogino, Y. Gene duplication of androgen receptor as an evolutionary driving force underlying the diversity of sexual characteristics in teleost fishes. Zool. Sci. 2024, 41, 68–76. [Google Scholar] [CrossRef]
  93. Jolly, C.; Katsiadaki, I.; Morris, S.; Le Belle, N.; Dufour, S.; Mayer, I.; Pottinger, T.G.; Scott, A.P. Detection of the anti-androgenic effect of endocrine disrupting environmental contaminants using in vivo and in vitro assays in the three-spined stickleback. Aquat. Toxicol. 2009, 92, 228–239. [Google Scholar] [CrossRef]
  94. Pang, Y.; Thomas, P. Role of G protein-coupled estrogen receptor 1 (GPER) in inhibition of oocyte maturation by endogenous estrogens in zebrafish. Dev. Biol. 2010, 342, 194–206. [Google Scholar] [CrossRef]
  95. Aizen, J.; Pang, Y.; Harris, C.; Converse, A.; Zhu, Y.; Aguirre, M.A.; Thomas, P. Roles of progesterone receptor membrane component 1 and membrane progestin receptor alpha in regulation of zebrafish oocyte maturation. Gen. Comp. Endocrinol. 2018, 263, 51–61. [Google Scholar] [CrossRef]
  96. He, L.; Shi, X.; Zeng, X.; Zhou, F.; Lan, T.; Chen, M.; Han, K. Characterization of the glucocorticoid receptor of large yellow croaker (Larimichthys crocea) and its expression in response to salinity and immune stressors. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2022, 265, 111124. [Google Scholar] [CrossRef]
  97. Bemanian, V.; Male, R.; Goksøyr, A. The aryl hydrocarbon receptor-mediated disruption of vitellogenin synthesis in the fish liver: Cross-talk between AhR- and ERα-signalling pathways. Comp. Hepatol. 2004, 3, 2. [Google Scholar] [CrossRef]
  98. Mortensen, A.S.; Arukwe, A. Interactions between estrogen- and Ah-receptor signalling pathways in primary culture of salmon hepatocytes exposed to nonylphenol and 3,3′,4,4′-tetrachlorobiphenyl (congener 77). Comp. Hepatol. 2007, 6, 2. [Google Scholar] [CrossRef]
  99. Mahoney, H.; Xie, Y.; Brinkmann, M.; Giesy, J.P. Next generation per- and poly-fluoroalkyl substances: Status and trends, aquatic toxicity, and risk assessment. Eco Environ. Health 2022, 1, 117–131. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, H.; Tan, J.T.; Emelyanov, A.; Korzh, V.; Gong, Z. Hepatic and extrahepatic expression of vitellogenin genes in the zebrafish, Danio rerio. Gene 2005, 356, 91–100. [Google Scholar] [CrossRef] [PubMed]
  101. Finn, R.N.; Kristoffersen, B.A. Vertebrate vitellogenin gene duplication in relation to the “3R hypothesis”: Correlation to the pelagic egg and the oceanic radiation of teleosts. PLoS ONE 2007, 2, e169. [Google Scholar] [CrossRef] [PubMed]
  102. Yilmaz, O.; Patinote, A.; Nguyen, T.; Com, E.; Pineau, C.; Bobe, J. Genome editing reveals reproductive and developmental dependencies on specific types of vitellogenin in zebrafish (Danio rerio). Mol. Reprod. Dev. 2019, 86, 1168–1188. [Google Scholar] [CrossRef]
  103. Trichet, V.; Buisine, N.; Mouchel, N.; Le Pennec, J.P.; Wolff, J. Genomic analysis of the vitellogenin locus in rainbow trout (Oncorhynchus mykiss) reveals a complex history of gene amplification and retroposon activity. Mol. Gen. Genet. 2000, 263, 828–837. [Google Scholar] [CrossRef]
  104. Carducci, F.; Biscotti, M.A.; Canapa, A. Vitellogenin gene family in vertebrates: Evolution and functions. Eur. Zool. J. 2019, 86, 233–240. [Google Scholar] [CrossRef]
  105. Finn, R.N.; Kolarevic, J.; Kongshaug, H.; Nilsen, F. Evolution and differential expression of a vertebrate vitellogenin gene cluster. BMC Evol. Biol. 2009, 9, 2. [Google Scholar] [CrossRef] [PubMed]
  106. Hiramatsu, N.; Matsubara, T.; Fujita, T.; Sullivan, C.V.; Hara, A. Multiple piscine vitellogenins: Biomarkers of fish exposure to estrogenic endocrine disruptors in aquatic environments. Mar. Biol. 2006, 149, 35–47. [Google Scholar] [CrossRef]
  107. Xiong, Z.; Yang, J.; Zhang, S.; Wang, Y.; Xu, S.; Guo, C.; Wang, D. Expression and localization of vitellogenin genes (VTG) and receptor (VGR) in the gonad development of silver pomfret Pampus argenteus. J. Oceanol. Limnol. 2023, 41, 1575–1592. [Google Scholar] [CrossRef]
  108. Yushko, L.V.; Shevlyakov, A.D.; Romazeva, M.A.; Medvedeva, A.V.; Sokolova, O.A. The Role of DNA Methylation in Zebrafish Models of CNS Diseases. J. Evol. Biochem. Physiol. 2024, 60, 973–987. [Google Scholar] [CrossRef]
  109. Kocerha, J.; Denslow, N.D. Steroidogenic acute regulatory protein in fish. J. Endocrinol. 2025, 265, e240232. [Google Scholar] [CrossRef]
  110. Wang, X.; Hill, D.; Tillitt, D.E.; Bhandari, R.K. Bisphenol A and 17α-ethinylestradiol-induced transgenerational differences in expression of osmoregulatory genes in the gill of medaka (Oryzias latipes). Aquat. Toxicol. 2019, 211, 227–234. [Google Scholar] [CrossRef]
  111. Xiong, S.; Ma, W.; Jing, J.; Zhang, J.; Dan, C.; Gui, J.F.; Mei, J. An miR-200 Cluster on Chromosome 23 Regulates Sperm Motility in Zebrafish. Endocrinology 2018, 159, 1982–1991. [Google Scholar] [CrossRef]
  112. Lee, J.; Kho, Y.; Kim, P.-G.; Ji, K. Exposure to bisphenol S alters the expression of microRNA in male zebrafish. Toxicol. Appl. Pharmacol. 2018, 338, 191–196. [Google Scholar] [CrossRef]
  113. Dourdin, T.S.; Rivière, G.; Cormier, A.; Di Poi, C.; Guyomard, K.; Rabiller, M.; Akcha, F.; Sadialiou, T.B.; Le Monier, P.; Sussarellu, R. Molecular and phenotypic effects of early exposure to an environmentally relevant pesticide mixture in the Pacific oyster, Crassostrea gigas. Environ. Pollut. 2023, 326, 121472. [Google Scholar] [CrossRef]
  114. Martyniuk, C.J.; Feswick, A.; Munkittrick, K.R.; Dreier, D.A.; Denslow, N.D. Twenty years of transcriptomics, 17α-ethinylestradiol, and fish. Gen. Comp. Endocrinol. 2020, 286, 113325. [Google Scholar] [CrossRef]
  115. Hemmer, M.J.; Bowman, C.J.; Hemmer, B.L.; Friedman, S.D.; Marcovich, D.; Kroll, K.J.; Denslow, N.D. Vitellogenin mRNA regulation and plasma clearance in male sheepshead minnows (Cyprinodon variegatus) after cessation of exposure to 17β-estradiol and p-nonylphenol. Aquat. Toxicol. 2002, 58, 99–112. [Google Scholar] [CrossRef]
  116. Geraudie, P.; Gerbron, M.; Hill, E.; Minier, C. Roach (Rutilus rutilus) reproductive cycle: A study of biochemical and histological parameters in a low contaminated site. Fish Physiol. Biochem. 2010, 36, 767–777. [Google Scholar] [CrossRef]
  117. Yadetie, F.; Karlsen, O.A.; Eide, M.; Hogstrand, C.; Goksøyr, A. Liver Transcriptome Analysis of Atlantic Cod (Gadus morhua) Exposed to PCB 153 Indicates Effects on Cell Cycle Regulation and Lipid Metabolism. BMC Genom. 2014, 15, 481. [Google Scholar] [CrossRef]
  118. Abd Elkader, H.-T.A.E.; Al-Shami, A.S. Unveiling the impact of bisphenol A on date mussels: Insights into oxidative stress, hormonal imbalance, gonadal atresia, and immune resilience. Mar. Environ. Res. 2025, 208, 107143. [Google Scholar] [CrossRef]
  119. Ji, C.; Wei, L.; Zhao, J.; Wu, H. Metabolomic analysis revealed that female mussel Mytilus galloprovincialis was sensitive to bisphenol A exposures. Environ. Toxicol. Pharmacol. 2014, 37, 844–849. [Google Scholar] [CrossRef] [PubMed]
  120. Ji, C.; Wu, H.; Wei, L.; Zhao, J.; Yu, J. Proteomic and metabolomic analysis reveal gender-specific responses of mussel Mytilus galloprovincialis to 2,2′,4,4′-tetrabromodiphenyl ether (BDE 47). Aquat. Toxicol. 2013, 140–141, 449–457. [Google Scholar] [CrossRef] [PubMed]
  121. Ankley, G.T.; Villeneuve, D.L. The fathead minnow in aquatic toxicology: Past, present and future. Aquat. Toxicol. 2006, 78, 91–102. [Google Scholar] [CrossRef]
  122. Ankley, G.T.; Bennett, R.S.; Erickson, R.J.; Hoff, D.J.; Hornung, M.W.; Johnson, R.D.; Mount, D.R.; Nichols, J.W.; Russom, C.L.; Schmieder, P.K.; et al. Adverse outcome pathways: A conceptual framework to support ecotoxicology research and risk assessment. Environ. Toxicol. Chem. 2010, 29, 730–741. [Google Scholar] [CrossRef] [PubMed]
  123. Yilmaz, O.; Patinote, A.; Com, E.; Pineau, C.; Bobe, J. Knock out of specific maternal vitellogenins in zebrafish (Danio rerio) evokes vital changes in egg proteomic profiles that resemble the phenotype of poor quality eggs. BMC Genom. 2021, 22, 308. [Google Scholar] [CrossRef]
  124. Zha, W.; Hu, W.; Ge, C.; Chen, J.; Cao, Z. Zebrafish as a model system for studying reproductive diseases. Front. Cell Dev. Biol. 2024, 12, 1481634. [Google Scholar] [CrossRef]
  125. Meng, X.; Bartholomew, C.; Craft, J.A. Differential expression of vitellogenin and oestrogen receptor genes in the liver of zebrafish, Danio rerio. Anal. Bioanal. Chem. 2010, 396, 625–630. [Google Scholar] [CrossRef] [PubMed]
  126. Dong, Y.; Wang, S.; Chen, J.; Zhang, Q.; Liu, Y.; You, C.; Monroig, Ó.; Tocher, D.R.; Li, Y. Hepatocyte Nuclear Factor 4α (HNF4α) Is a Transcription Factor of Vertebrate Fatty Acyl Desaturase Gene as Identified in Marine Teleost Siganus canaliculatus. PLoS ONE 2016, 11, e0160361. [Google Scholar] [CrossRef] [PubMed]
  127. Maglich, J.M.; Caravella, J.A.; Lambert, M.H.; Willson, T.M.; Moore, J.T.; Ramamurthy, L. The First Completed Genome Sequence from a Teleost Fish (Fugu rubripes) Adds Significant Diversity to the Nuclear Receptor Superfamily. Nucleic Acids Res. 2003, 31, 4051–4058. [Google Scholar] [CrossRef] [PubMed]
  128. Bainy, A.C.D.; Kubota, A.; Goldstone, J.V.; Lille-Langøy, R.; Karchner, S.I.; Celander, M.C.; Hahn, M.E.; Goksøyr, A.; Stegeman, J.J. Functional Characterization of a Full-Length Pregnane X Receptor, Expression in vivo, and Identification of PXR Alleles in Zebrafish (Danio rerio). Aquat. Toxicol. 2013, 142–143, 447–457. [Google Scholar] [CrossRef]
  129. Brion, F.; Nilsen, B.M.; Eidem, J.K.; Goksøyr, A.; Porcher, J.M. Development and validation of an enzyme-linked immunosorbent assay to measure vitellogenin in the zebrafish (Danio rerio). Environ. Toxicol. Chem. 2002, 21, 1699–1708. [Google Scholar] [CrossRef]
  130. Jobling, S.; Nolan, M.; Tyler, C.R.; Brighty, G.; Sumpter, J.P. Widespread sexual disruption in wild fish. Environ. Sci. Technol. 1998, 32, 2498–2506. [Google Scholar] [CrossRef]
  131. Markell, L.K.; Mingoia, R.T.; Peterson, H.M.; Yao, J.; Waters, S.M.; Finn, J.P.; Nabb, D.L.; Han, X. Endocrine disruption screening by protein and gene expression of vitellogenin in freshly isolated and cryopreserved rainbow trout hepatocytes. Chem. Res. Toxicol. 2014, 27, 1450–1457. [Google Scholar] [CrossRef]
  132. Gräns, J.; Wassmur, B.; Celander, M.C. One-way inhibiting cross-talk between arylhydrocarbon receptor (AhR) and estrogen receptor (ER) signaling in primary cultures of rainbow trout hepatocytes. Aquat. Toxicol. 2010, 100, 263–270. [Google Scholar] [CrossRef]
  133. Ricciardi, F.; Matozzo, V.; Marin, M.G. Effects of 4-nonylphenol exposure in mussels (Mytilus galloprovincialis) and crabs (Carcinus aestuarii) with particular emphasis on vitellogenin induction. Mar. Pollut. Bull. 2008, 57, 365–372. [Google Scholar] [CrossRef]
  134. Nolte, T.M.; De Cooman, W.; Vink, J.P.M.; Elst, R.; Ryken, E.; Ragas, A.M.J.; Hendriks, A.J. Bioconcentration of Organotin Cations during Molting Inhibits Heterocypris incongruens Growth. Environ. Sci. Technol. 2020, 54, 14288–14301. [Google Scholar] [CrossRef]
  135. Gong, J.; Li, J.; Tian, X.; Zeng, B.; Wang, B.; Yu, F. The Retinoid X Receptor from Mud Crab: New Insights into Its Role in Vitellogenesis. Sci. Rep. 2016, 6, 23654. [Google Scholar] [CrossRef] [PubMed]
  136. Fenske, M.; van Aerle, R.; Brack, S.; Tyler, C.R.; Segner, H. Development and validation of a homologous zebrafish vitellogenin ELISA and its application for studies on estrogenic chemicals. Comp. Biochem. Physiol. C Toxicol. Pharmacol. 2001, 129, 217–232. [Google Scholar] [CrossRef] [PubMed]
  137. Barucca, M.; Canapa, A.; Olmo, E.; Regoli, F. Analysis of vitellogenin gene induction as a valuable biomarker of estrogenic exposure in various Mediterranean fish species. Environ. Res. 2006, 101, 68–73. [Google Scholar] [CrossRef] [PubMed]
  138. Mukherjee, U.; Maitra, S. Impact of metabolic disrupting chemicals on redox homeostasis, energy sensors, receptor modulation, and hormone metabolism: A comparative account in teleost and mammalian model organisms. Aquac. Fish. 2024, 9, 455–485. [Google Scholar] [CrossRef]
  139. Kinnberg, K.; Toft, G. Effects of estrogenic and antiandrogenic compounds on the testis structure of adult male guppies (Poecilia reticulata). Ecotoxicol. Environ. Saf. 2003, 54, 16–24. [Google Scholar] [CrossRef]
  140. Levi, L.; Pekarski, I.; Gutman, E.; Fortina, P.; Hyslop, T.; Biran, J.; Levavi-Sivan, B.; Lubzens, E. Revealing genes associated with vitellogenesis in the liver of the zebrafish (Danio rerio) by transcriptome profiling. BMC Genom. 2009, 10, 141. [Google Scholar] [CrossRef]
  141. Dorts, J.; Richter, C.A.; Wright-Osment, M.K.; Ellersieck, M.R.; Carter, B.J.; Tillitt, D.E. The genomic transcriptional response of female fathead minnows (Pimephales promelas) to an acute exposure to the androgen, 17β-trenbolone. Aquat. Toxicol. 2009, 91, 44–53. [Google Scholar] [CrossRef]
  142. Rios-Sicairos, J.; Betancourt-Lozano, M.; Leal-Tarin, B.; Hernandez-Cornejo, R.; Aguilar-Zarate, G.; Garcia-De-La-Parra, L.M.; Gutierrez, J.N.; Marquez-Rocha, F.; Garcia-Gasca, A. Heat-shock protein (Hsp70) and cytochrome P-450 (CYP1A) in the white mullet Mugil curema (Pisces: Mugilidae) as biomarkers to assess environmental quality in coastal lagoons. J. Environ. Sci. Health A Tox. Hazard. Subst. Environ. Eng. 2010, 45, 68–74. [Google Scholar] [CrossRef]
  143. Kolli, R.T.; Glenn, T.C.; Bringolf, R.B.; Henderson, M.; Cummings, B.S.; Kenneke, J.F. Changes in CpG Methylation of the vtg1 Promoter in Adult Male Zebrafish after Exposure to 17α-Ethinylestradiol. Environ. Toxicol. Chem. 2024, 43, 1547–1556. [Google Scholar] [CrossRef]
  144. Borcier, E.; Artigaud, S.; Gaillard, J.C.; Armengaud, J.; Charrier, G.; Couteau, J.; Receveur, J.; Ouddane, B.; Diop, M.; Amara, R.; et al. Coupling caging and proteomics on the European flounder (Platichthys flesus) to assess the estuarine water quality at micro scale. Sci. Total Environ. 2019, 695, 133760. [Google Scholar] [CrossRef]
  145. See, M.J.; Bencic, D.C.; Flick, R.W.; Lazorchak, J.; Biales, A.D. Characterization of vitellogenin concentration in male fathead minnow mucus compared to plasma and liver mRNA. Ecotoxicol. Environ. Saf. 2022, 236, 113428. [Google Scholar] [CrossRef] [PubMed]
  146. Hennies, M.; Wiesmann, M.; Allner, B.; Sauerwein, H. Vitellogenin in carp (Cyprinus carpio) and perch (Perca fluviatilis): Purification, characterization and development of an ELISA for the detection of estrogenic effects. Sci. Total Environ. 2003, 309, 93–103. [Google Scholar] [CrossRef] [PubMed]
  147. Bhat, R.A.; Singh, R.; Pandey, A.; Chauhan, R.S. Role of Sex-Biased miRNAs in Teleosts—A Review. Aquac. Res. 2021, 52, 5071–5086. [Google Scholar] [CrossRef]
  148. Hamilton, P.B.; Baynes, A.; Nicol, E.; Harris, G.; Uren Webster, T.M.; Beresford, N.; Straszkiewicz, M.; Jobling, S.; Tyler, C.R. Feminizing effects of ethinylestradiol in roach (Rutilus rutilus) populations with different estrogenic pollution exposure histories. Aquat. Toxicol. 2022, 249, 106229. [Google Scholar] [CrossRef]
  149. Baynes, A.; Lange, A.; Beresford, N.; Bryden, E.; Whitlock, K.; Tyler, C.R.; Jobling, S. Endocrine Disruption Is Reduced but Still Widespread in Wild Roach (Rutilus rutilus) Living in English Rivers. Environ. Sci. Technol. 2023, 57, 12632–12641. [Google Scholar] [CrossRef]
  150. Kirby, M.F.; Allen, Y.T.; Dyer, R.A.; Feist, S.W.; Katsiadaki, I.; Matthiessen, P.; Scott, A.P.; Smith, A.; Stentiford, G.D.; Thain, J.E.; et al. Surveys of Plasma Vitellogenin and Intersex in Male Flounder (Platichthys flesus) as Measures of Endocrine Disruption in UK Estuaries: Temporal Trends, 1996–2001. Environ. Toxicol. Chem. 2004, 23, 748–758. [Google Scholar] [CrossRef]
  151. George, S.; Gubbins, M.; MacIntosh, A.; Reynolds, W.; Sabine, V.; Scott, A.; Thain, J. A comparison of pollutant biomarker responses with transcriptional responses in European flounders (Platichthys flesus) subjected to estuarine pollution. Mar. Environ. Res. 2004, 58, 571–575. [Google Scholar] [CrossRef]
  152. Ríos-Sicairos, J.; García-Rodríguez, F.; Páez-Osuna, F.; Sánchez-Como, J.; Acosta-Vargas, B.; Frías-Espericueta, M.G. HSP70 and CYP1A in White Mullet (Mugil curema) as Biomarkers to Assess Environmental Quality in Coastal Lagoons. Mar. Environ. Res. 2010, 70, 11–16. [Google Scholar] [CrossRef]
  153. Cocci, P.; Mosconi, G.; Palermo, F.A. An In Silico and In Vitro Study for Investigating Estrogenic Endocrine Effects of Emerging Persistent Pollutants Using Primary Hepatocytes from Grey Mullet (Mugil cephalus). Environments 2021, 8, 58. [Google Scholar] [CrossRef]
  154. Ramos-Júdez, S.; Estévez, A.; González-López, W.Á.; Duncan, N. Lipid and fatty acid composition of muscle, liver, ovary, and peritoneal fat in wild flathead grey mullet (Mugil cephalus) according to ovarian development. Theriogenology 2023, 198, 317–326. [Google Scholar] [CrossRef]
  155. Ortiz-Zarragoitia, M.; Bizarro, C.; Rojo-Bartolomé, I.; Diaz de Cerio, O.; Cajaraville, M.P.; Cancio, I. Mugilid Fish Are Sentinels of Exposure to Endocrine Disrupting Compounds in Coastal and Estuarine Environments. Mar. Drugs 2014, 12, 4756–4782. [Google Scholar] [CrossRef] [PubMed]
  156. Anastasiadi, D.; Vandeputte, M.; Sánchez-Baizán, N.; Allal, F.; Piferrer, F. Dynamic epimarks in sex-related genes predict gonad phenotype in the European sea bass, a fish with mixed genetic and environmental sex determination. Epigenetics 2018, 13, 988–1011. [Google Scholar] [CrossRef] [PubMed]
  157. Yadetie, F.; Zhang, X.; Hanna, E.M.; Aranguren-Abadía, L.; Eide, M.; Blaser, N.; Brun, M.; Jonassen, I.; Goksøyr, A.; Karlsen, O.A. RNA-Seq Analysis of Atlantic Cod Liver Slices Exposed to Benzo[a]pyrene and 17α-Ethinylestradiol. Aquat. Toxicol. 2018, 201, 174–186. [Google Scholar] [CrossRef]
  158. Li, B.; Zhang, Y.; Du, J.; Liu, C.; Zhou, G.; Li, M.; Yan, Z. Application of Multi-Omics Techniques in Aquatic Ecotoxicology: A Review. Toxics 2025, 13, 653. [Google Scholar] [CrossRef] [PubMed]
  159. Ankley, G.T.; Blackwell, B.R.; Cavallin, J.E.; Doering, J.A.; Feifarek, D.J.; Jensen, K.M.; Kahl, M.D.; LaLone, C.A.; Poole, S.T.; Randolph, E.C.; et al. Adverse outcome pathway network-based assessment of the interactive effects of an androgen receptor agonist and an aromatase inhibitor on fish endocrine function. Environ. Toxicol. Chem. 2020, 39, 913–922. [Google Scholar] [CrossRef]
  160. Valdivieso, A.; Anastasiadi, D.; Ribas, L.; Piferrer, F. Development of Epigenetic Biomarkers for the Identification of Sex and Thermal Stress in Fish Using DNA Methylation Analysis and Machine Learning Procedures. Mol. Ecol. Resour. 2023, 23, 453–470. [Google Scholar] [CrossRef]
  161. Cannea, F.B.; Follesa, M.C.; Porcu, C.; Rossino, R.; Olianas, A.; Rescigno, A.; Padiglia, A. Antibodies targeting the European lobster (Palinurus elephas) vitellogenin developed by mRNA isolation and in-silico-designed antigenic peptides. Biol. Open 2022, 11, bio059019. [Google Scholar] [CrossRef]
  162. De Wit, M.; Keil, D.; van der Ven, K.; Vandamme, S.; Witters, E.; De Coen, W. An integrated transcriptomic and proteomic approach characterizing estrogenic and metabolic effects of 17α-ethinylestradiol in zebrafish (Danio rerio). Gen. Comp. Endocrinol. 2010, 167, 190–201. [Google Scholar] [CrossRef]
  163. Zhao, Z.; Zhao, Q.; Wang, H.; Wei, L.; Wang, S.; Li, S.; Yuan, D.; Wang, Z. Integrated transcriptomic and metabolomic analyses identify key factors in the vitellogenesis of juvenile Sichuan bream (Sinibrama taeniatus). Front. Mar. Sci. 2023, 10, 1243767. [Google Scholar] [CrossRef]
  164. Pinto, P.I.; Anjos, L.; Estêvão, M.D.; Santos, S.; Santa, C.; Manadas, B.; Monsinjon, T.; Canário, A.V.M.; Power, D.M. Proteomics of sea bass skin-scales exposed to the emerging pollutant fluoxetine compared to estradiol. Sci. Total Environ. 2022, 814, 152671. [Google Scholar] [CrossRef]
  165. Frelih, M.; Ayobahan, S.U.; Marghany, F.; Essfeld, F.; Eilebrecht, S. Toxicogenomic signatures of estrogen-related modes of action in the zebrafish embryo. Environ. Toxicol. Chem. 2025, 44, 2568–2579. [Google Scholar] [CrossRef] [PubMed]
  166. Villeneuve, D.L.; Bush, K.; Hazemi, M.; Hoang, J.X.; Le, M.; Blackwell, B.R.; Stacy, E.; Flynn, K.M. Derivation of Transcriptomics-Based Points of Departure for 20 Per- or Polyfluoroalkyl Substances Using a Larval Fathead Minnow (Pimephales promelas) Reduced Transcriptome Assay. Environ. Toxicol. Chem. 2024, 44, 2455–2469. [Google Scholar] [CrossRef] [PubMed]
  167. Flynn, K.M.; Le, M.; Hazemi, M.; Biales, A.; Bencic, D.C.; Blackwell, B.R.; Bush, K.; Flick, R.; Hoang, J.X.; Martinson, J.; et al. Comparing Transcriptomic Points of Departure to Apical Effect Concentrations for Larval Fathead Minnow Exposed to Chemicals with Four Different Modes of Action. Arch. Environ. Contam. Toxicol. 2024, 86, 346–362. [Google Scholar] [CrossRef] [PubMed]
  168. Villeneuve, D.L.; Le, M.; Hazemi, M.; Biales, A.; Bencic, D.C.; Bush, K.; Flick, R.; Martinson, J.; Morshead, M.; Rodriguez, K.S.; et al. Pilot Testing and Optimization of a Larval Fathead Minnow High-Throughput Transcriptomics Assay. Curr. Res. Toxicol. 2022, 4, 100099. [Google Scholar] [CrossRef]
  169. O’Brien, J.; Mitchell, C.; Auerbach, S.; Doonan, L.; Ewald, J.; Everett, L.; Faranda, A.; Johnson, K.; Reardon, A.; Rooney, J.; et al. Bioinformatic Workflows for Deriving Transcriptomic Points of Departure: Current Status, Data Gaps, and Research Priorities. Toxicol. Sci. 2025, 203, 147–159. [Google Scholar] [CrossRef]
  170. Gasser, L.; Schür, C.; Perez-Cruz, F.; Schirmer, K.; Baity-Jesi, M. Machine Learning-Based Prediction of Fish Acute Mortality: Implementation, Interpretation, and Regulatory Relevance. Environ. Sci. Adv. 2024, 3, 1124–1138. [Google Scholar] [CrossRef]
  171. Nam, S.-E.; Kim, J.-H.; Kim, H.-J.; Park, S.; Lee, S.; Kim, B.-M.; Park, J.-C. Multi-Omics Integration Reveals Underlying Toxicological Mechanisms of Triclosan in the Freshwater Fish Zacco platypus. Aquat. Toxicol. 2025, 287, 107522. [Google Scholar] [CrossRef]
  172. Whitehead, A.; Pilcher, W.; Champlin, D.; Nacci, D. Common mechanism underlies repeated evolution of extreme pollution tolerance. Proc. R. Soc. B 2012, 279, 427–433. [Google Scholar] [CrossRef]
  173. Whitehead, A.; Clark, B.W.; Reid, N.M.; Hahn, M.E.; Nacci, D. When evolution is the solution to pollution: Key principles, and lessons from rapid repeated adaptation of killifish (Fundulus heteroclitus) populations. Evol. Appl. 2017, 10, 762–783. [Google Scholar] [CrossRef]
  174. Ajisafe, O.M.; Adekunle, Y.A.; Egbon, E.; Ogbonna, C.E.; Olawade, D.B. The role of machine learning in predictive toxicology: A review of current trends and future perspectives. Life Sci. 2025, 378, 123821. [Google Scholar] [CrossRef]
  175. Conolly, R.B.; Ankley, G.T.; Cheng, W.; Mayo, M.L.; Miller, D.H.; Perkins, E.J.; Villeneuve, D.L.; Watanabe, K.H. Quantitative adverse outcome pathways and their application to predictive toxicology. Environ. Sci. Technol. 2017, 51, 4661–4672. [Google Scholar] [CrossRef]
  176. Piccardo, M.; Vellani, V.; Anselmi, S.; Grazioli, E.; Renzi, M.; Terlizzi, A.; Pittura, L.; D’Errico, G.; Regoli, F.; Bevilacqua, S. The application of the weight-of-evidence approach for an integrated ecological risk assessment of marine protected sites. Ecol. Indic. 2024, 159, 111676. [Google Scholar] [CrossRef]
  177. OECD. Endocrine Disrupting Chemicals in Freshwater: Monitoring and Regulating Water Quality; OECD Studies on Water; OECD Publishing: Paris, France, 2023. [Google Scholar] [CrossRef]
  178. Wernersson, A.S.; Carere, M.; Maggi, C.; Tusil, P.; Soldan, P.; James, A.; Sanchez, W.; Dulio, V.; Broeg, K.; Reifferscheid, G.; et al. The European technical report on aquatic effect-based monitoring tools under the Water Framework Directive. Environ. Sci. Eur. 2015, 27, 7. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of sex-specific effects of xenoestrogens (XEs) on VTG expression in fish. Under normal conditions, males do not express VTG, whereas XE exposure induces vtg transcription and ectopic VTG synthesis. In females, where VTG is physiologically expressed, XEs enhance vtg upregulation and hepatic VTG synthesis. VTG is secreted into the bloodstream, transported to the ovary, and internalized by oocytes via vitellogenin receptors (VGRs). Examples of protein detection methods are shown (Western blot and ELISA); additional approaches are described in the text. Arrows indicate process direction; ↑ denotes vtg upregulation.
Figure 1. Schematic representation of sex-specific effects of xenoestrogens (XEs) on VTG expression in fish. Under normal conditions, males do not express VTG, whereas XE exposure induces vtg transcription and ectopic VTG synthesis. In females, where VTG is physiologically expressed, XEs enhance vtg upregulation and hepatic VTG synthesis. VTG is secreted into the bloodstream, transported to the ovary, and internalized by oocytes via vitellogenin receptors (VGRs). Examples of protein detection methods are shown (Western blot and ELISA); additional approaches are described in the text. Arrows indicate process direction; ↑ denotes vtg upregulation.
Genes 16 01317 g001
Table 1. Representative examples of endocrine disruption in aquatic taxa. The table includes freshwater and marine fish, mollusks, and crustaceans, summarizing key EDCs, their biological effects, and sex-specific responses.
Table 1. Representative examples of endocrine disruption in aquatic taxa. The table includes freshwater and marine fish, mollusks, and crustaceans, summarizing key EDCs, their biological effects, and sex-specific responses.
Taxonomic GroupEDCsMain EffectsSex-Specific EffectsRepresentative References
Fish (P. promelas)17α-ethinylestradiol (EE2)VTG induction, intersex development, population collapse in whole-lake experimentMales strongly induced; females affected at reproductive stages[38,39]
Fish (R. rutilus)Nonylphenol, wastewater effluentsVTG induction, intersex gonads, feminization of wild populationsFeminization in males; females within physiological baseline[34,35]
Fish (various freshwater species)AtrazineAromatase induction, altered sex steroid balance, feminizationMales overexpress aromatase; feminization observed[32,33]
Fish (G. morhua)17α-ethinylestradiol (EE2)Hepatic reprogramming of lipid metabolism and immune pathways (multi-omics)Sex-biased transcriptional shifts, stronger in males[76]
Fish (P. flesus)Field exposure in polluted estuariesCo-induction of VTG, choriogenins, HSP70/90; evidence of estrogenic disruptionMale induction of VTG/Chg; females less responsive[77]
Mollusks (Mytilus spp.)Bisphenol A (BPA), EE2Disrupted embryogenesis, altered lipid metabolism and vitellogenesis; inconsistent VTG-like inductionSex-dependent responses unclear; variable results among studies[36,37,40,41,71]
Mollusks (Crassostrea gigas)Xenoestrogens (e.g., BPA, EE2)Altered DNA methylation and non-coding RNAs; disruption of gametogenesisEpigenetic changes may affect gametes differently between sexes[45]
Crustaceans (general, e.g., crabs, amphipods)Organotins, pesticidesDisruption of EcR/RXR signaling; impaired molting, vitellogenesis, and reproductionSex-dependent reproductive impairment observed[79,80,81,82,83]
Crustaceans (Marsupenaeus japonicus)Methyl farnesoate (MF), 20-hydroxyecdysone (20E)Experimental exposure confirmed disruption of molting and reproduction (EcR/RXR axis)Disruption of hormone balance affects both sexes differently[84]
Table 2. Key genes and regulatory elements affected by EDCs in fish and aquatic invertebrates, with associated functions, mechanisms, and sex-specific responses where known. Arrows (→) indicate the direction of molecular or physiological changes.
Table 2. Key genes and regulatory elements affected by EDCs in fish and aquatic invertebrates, with associated functions, mechanisms, and sex-specific responses where known. Arrows (→) indicate the direction of molecular or physiological changes.
GeneGene Product and FunctionMain EDCsRegulation by EDCsMolecular MechanismSex-Specific ResponsesReferences
vtg (D. rerio, salmonids, R. rutilus)VTG (yolk precursor protein)EE2, BPA, NPStrongly upregulatedERα → EREs; promoter hypomethylation enhances inductionNegligible baseline in males → strong induction under EDCs; physiological in females[100,101,102,103,104,105,106,107]
zp2, zp3 (teleosts)ZP2/ZP3 (zona pellucida proteins, egg envelope)EE2, BPAUpregulatedER-dependent transcription; co-regulated with vtgFemale-biased; weak/no expression in males under control conditions[88,90]
chg (teleosts)Chg (choriogenins, egg envelope glycoproteins)EE2, NPUpregulatedERα-mediated transcriptionStrong biomarker in males (ectopic induction)[106,107]
cyp19a1a (teleosts, amphibians)CYP19A1A (Aromatase A, estrogen synthesis)Atrazine, BPAInducedSteroidogenesis; conversion androgens → estrogensOverexpression in males → feminization[91,110]
cyp1a (teleosts, invertebrates)CYP1A (cytochrome P450 1A, detoxification enzyme)Dioxins, PCBsUpregulatedAhR/ARNT → XREs; antagonizes ER signalingSuppression of vtg in males[97,98]
star (teleosts)StAR (steroidogenic acute regulatory protein, cholesterol transport)BPA, metalsAlteredSteroid hormone biosynthesisAffects sex steroid balance in both sexes[109]
hsd family (teleosts)HSDs (hydroxysteroid dehydrogenases, steroid metabolism)BPA, metalsModulatedSteroidogenesisKey in androgen/estrogen ratio; dysregulation impacts fertility[91,110]
dnmt (teleosts, mollusks)DNMTs (DNA methyltransferases, epigenetic regulators)BPA, EE2AlteredDNA methylation changes at promoters (vtg1, cyp19a1a)Sex-specific promoter methylation (e.g., vtg1 hypo in females, hyper in males)[108,113]
miR-200 family (D. rerio)miR-200 family (epigenetic regulators of spermatogenesis)EE2Upregulatedp53–miR-200 axisReduced sperm motility in males[111,112]
vtg-like (bivalves, e.g., Mytilus spp.)VTG-like yolk precursor proteinsBPA, EE2Inconsistent inductionEstrogenic signaling vs. oxidative stress cross-talkVariable; induction sometimes absent in males/females[118,119,120]
EcR/RXR (crustaceans)Ecdysone receptor/Retinoid X receptor (molting, reproduction)Organotins, pesticidesDisruptedInterference with 20E–MF axisImpaired molting and vitellogenesis; sex-dependent reproductive impairment[92,93,121,122]
Table 3. VTG genes and paralogs in different taxa, their physiological induction in females, induction under EDCs in males and juveniles, main regulatory pathways, and relevant notes.
Table 3. VTG genes and paralogs in different taxa, their physiological induction in females, induction under EDCs in males and juveniles, main regulatory pathways, and relevant notes.
VTG Genes and Paralogs (Species)Physiological
Induction (Females)
Induction Under EDC Exposure (Males and Juveniles)Main Regulatory PathwayNotesReferences
vtgAa, vtgAb
(D. rerio)
Strong induction during vitellogenesis under estrogen controlRobust induction by EE2, BPA, NonylphenolERα binding EREs; promoter demethylationEssential for oocyte maturation and fertility[100,102,123,125,129,130,137]
vtgC
(D. rerio)
Weakly responsive in femalesMinimal induction under EDCsWeak ERα responsivenessSecondary role; subfunctionalized paralog[101,104,105]
multiple vtg paralogs (salmonids, e.g., Salmo salar)Several paralogs highly induced; others weakDifferential induction depending on paralogERα/EREs; subfunctionalizationLarge gene arrays due to genome duplication[103,105,106]
VTG-like proteins
(Mytilus spp.)
Baseline expression variableSometimes induced by BPA, Nonylphenol, EE2 (inconsistent)ER-related transcripts; stress pathwaysBiomarker value debated, not consistent[118,119,120,133]
VTG-like transcripts (crustaceans, e.g., crabs, shrimps)VTG expression increases during female vitellogenesis under hormonal controlLimited induction after organotin and other EDC exposure; responses vary across taxaRXR/EcR pathways; interaction with methyl farnesoate and 20-hydroxyecdysoneVTG investigated as a potential biomarker in crustaceans, but responses remain variable across taxa[21,84,85,135]
Table 4. Sex-specific biomarkers associated with endocrine disruption in aquatic organisms. The table integrates evidence from fish, mollusks, and crustaceans, summarizing biomarker responses in males and females, detection methods, and their ecotoxicological relevance.
Table 4. Sex-specific biomarkers associated with endocrine disruption in aquatic organisms. The table integrates evidence from fish, mollusks, and crustaceans, summarizing biomarker responses in males and females, detection methods, and their ecotoxicological relevance.
BiomarkerSpecies and TaxaResponse in MalesResponse in FemalesAssayEcotoxicological RelevanceReferences
VTGFish (Cyprinis,
Salmonids)
Strong inductionBaseline fluctuationELISA, qPCR, Western blotSensitive biomarker of estrogenic exposure[15,16,17,18,106,136,137,146]
ERα/ERβ isoformsTeleostsDifferential modulationVariable (cycle-dependent)qPCR, RNA-SeqKey regulators of VTG and reproduction[88,89,90,125]
miR-200 clusterD. rerioReduced sperm motilityNot determinedqPCR, small RNA-seqEpigenetic marker of fertility disruption[111,112,147]
DNMTs/ncRNAsMollusks (Mytilus, Crassostrea)Altered methylation, disrupted ncRNAsGametogenesis impairmentqPCR, methylome analysisEpigenetic biomarkers of endocrine disruption in invertebrates[20,36,37,113]
EcR/RXR signalingCrustaceans (crabs, amphipods, shrimps)Impaired molting and reproductionAltered vitellogenesisTranscriptomics, receptor assaysBiomarkers of endocrine disruption via the MF/20E hormonal pathway[21,60,84,85,135]
Table 5. Omics approaches in aquatic ecotoxicology of endocrine disruptors. The table summarizes transcriptomic, proteomic, metabolomic, and epigenomic strategies, highlighting their main advantages, limitations, representative case studies, and relevance to sex-specific responses and VTG regulation.
Table 5. Omics approaches in aquatic ecotoxicology of endocrine disruptors. The table summarizes transcriptomic, proteomic, metabolomic, and epigenomic strategies, highlighting their main advantages, limitations, representative case studies, and relevance to sex-specific responses and VTG regulation.
Omics ApproachAdvantagesLimitationsRepresentative
Examples
Relevance to
Sex-Specific
Responses and VTG
References
TranscriptomicsGenome-wide sensitivity; reveals estrogenic signatures; identifies co-regulated pathwaysHigh data complexity; requires bioinformatic pipelinesEE2—D. rerio (estrogenic signatures); RNA-Seq—G. morhua (immune/metabolic reprogramming); M. edulis (biomarkers of endocrine disruption)Sex-biased regulation of vtg, zp2/3, chg; stronger shifts in males due to lower baseline estrogen[24,117,148,149]
ProteomicsLinks directly to protein biomarkers; detects post-translational modificationsRequires species-specific antibodies; less standardized across taxaMale C. variegatus—hepatic remodeling under EE2; P. flesus (field proteomics: VTG, Chg, HSP70/90); M. galloprovincialis (oxidative stress)Confirms VTG isoform induction; links sex-specific stress to protein remodeling[11,12,120,140,141]
MetabolomicsCaptures functional metabolic shifts; sensitive to physiological disruptionLower mechanistic specificity; metabolites can be transientR. rutilus—EE2 reduced circulating steroids; G. morhua (multi-omics: systemic reprogramming); mussels (BDE-47/TBBPA: altered energy metabolism)Sex-dependent metabolic trade-offs; VTG synthesis linked to lipid/energy metabolism[24,25,119]
EpigenomicsReveals persistent and transgenerational effects; potential biomarkersLimited cross-species data; methods still emerging in ecotoxD. reriovtg1 promoter methylation under EE2; O. latipes—methylation shifts (BPA/EE2); M. galloprovincialis—altered dnmt; C. gigas—ncRNA disruptionExplains sex-specific sensitivity (promoter methylation differences); reveals heritable effects beyond VTG induction[10,73,111,112,143]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cannea, F.B.; Porcu, C.; Follesa, M.C.; Padiglia, A. Sex-Specific Molecular and Genomic Responses to Endocrine Disruptors in Aquatic Species: The Central Role of Vitellogenin. Genes 2025, 16, 1317. https://doi.org/10.3390/genes16111317

AMA Style

Cannea FB, Porcu C, Follesa MC, Padiglia A. Sex-Specific Molecular and Genomic Responses to Endocrine Disruptors in Aquatic Species: The Central Role of Vitellogenin. Genes. 2025; 16(11):1317. https://doi.org/10.3390/genes16111317

Chicago/Turabian Style

Cannea, Faustina Barbara, Cristina Porcu, Maria Cristina Follesa, and Alessandra Padiglia. 2025. "Sex-Specific Molecular and Genomic Responses to Endocrine Disruptors in Aquatic Species: The Central Role of Vitellogenin" Genes 16, no. 11: 1317. https://doi.org/10.3390/genes16111317

APA Style

Cannea, F. B., Porcu, C., Follesa, M. C., & Padiglia, A. (2025). Sex-Specific Molecular and Genomic Responses to Endocrine Disruptors in Aquatic Species: The Central Role of Vitellogenin. Genes, 16(11), 1317. https://doi.org/10.3390/genes16111317

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop